EP2559698A2 - Mutante doppelt zyklierte Rezeptorpeptide zur Hemmung von beta-1-Adrenozeptorantikörper - Google Patents

Mutante doppelt zyklierte Rezeptorpeptide zur Hemmung von beta-1-Adrenozeptorantikörper Download PDF

Info

Publication number
EP2559698A2
EP2559698A2 EP12178671A EP12178671A EP2559698A2 EP 2559698 A2 EP2559698 A2 EP 2559698A2 EP 12178671 A EP12178671 A EP 12178671A EP 12178671 A EP12178671 A EP 12178671A EP 2559698 A2 EP2559698 A2 EP 2559698A2
Authority
EP
European Patent Office
Prior art keywords
cys
amino acid
cyclic peptide
ser
peptide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP12178671A
Other languages
English (en)
French (fr)
Other versions
EP2559698A3 (de
EP2559698B1 (de
Inventor
Roland Jahns
Valérie JAHNS
Martin Lohse
Viacheslav Nikolaev
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Julius Maximilians Universitaet Wuerzburg
Original Assignee
Julius Maximilians Universitaet Wuerzburg
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Julius Maximilians Universitaet Wuerzburg filed Critical Julius Maximilians Universitaet Wuerzburg
Priority to EP12178671.9A priority Critical patent/EP2559698B1/de
Publication of EP2559698A2 publication Critical patent/EP2559698A2/de
Publication of EP2559698A3 publication Critical patent/EP2559698A3/de
Application granted granted Critical
Publication of EP2559698B1 publication Critical patent/EP2559698B1/de
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/64Cyclic peptides containing only normal peptide links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to novel ⁇ -AR homologous cyclopeptide-mutants comprising only two cysteine residues able to form an intramolecular linkage, to iinear peptides that can form these cyclopeptitde-mutants and to nucleic acid molecules encoding these cyclopeptide-mutants and linear peptides.
  • vectors and recombinant host cells comprising said nucleic acid molecule and a method for producing the disclosed cyclopeptide-mutants are provided.
  • a composition comprising the peptides, nucleic acid molecules, vectors or host cells of the invention.
  • the present invention also relates to therapeutic and diagnostic means, methods and uses taking advantage of the peptides of the invention and to means, methods and uses for detecting anti- ⁇ -adrenergic receptor antibodies like anti- ⁇ 1 -adrenergic receptor antibodies.
  • DCM dilated cardiomyopathy
  • an initial acute inflammatory reaction may proceed into a kind of low-grade inflammation (MacLellan 2003) facilitating the development of abnormal or misled immune responses to the primary infectious trigger (Freedman 2004,de 2005, MacLellan and Lusis 2003, Maekawa 2007, Smulski 2006).
  • ⁇ 1 -adrenergic receptor results in generation of cross-reacting antibodies in about 30% of the Chagas' patients (Ferrari 1995). Because receptor-autoantibodies from patients with DCM preferentially recognize the C-terminal half of the same loop (Wallukat 1995), it was speculated that these antibodies might originate from molecular mimicry between the ⁇ 1 -AR and a hitherto unidentified viral pathogen (Magnusson 1996).
  • Another -probably more relevant- mechanism leading to the production of endogenous cardiac autoantibodies would be primary cardiac injury followed by (sudden or chronic) liberation of a "critical amount" of antigenic determinants from the myocyte membrane or cytoplasm, previously hidden to the immune system.
  • Such injury most likely occurs upon acute infectious (myocarditis), toxic, or ischemic heart disease (myocardial infarction) resulting in myocyte apoptosis or necrosis (Caforio 2002, Rose 2001).
  • autoimmune response may then induce an autoimmune response, which in the worst case results in perpetuation of immune-mediated myocyte damage involving either cellular (e.g., T-cell), or humoral (e.g., B-cell) immune responses, or co-activation of both the innate and the adaptive immune system (Eriksson 2003, Rose 2001).
  • cellular e.g., T-cell
  • humoral e.g., B-cell
  • anti-muscarinic antibodies exhibiting an agonist-like action on the cardiac M2 acetylcholine-receptor
  • agonistic anti- ⁇ 1 -AR antibodies have been associated with both the occurrence of severe arrhythmia at the ventricular level (Christ 2001, Iwata 2001 a), and the development of (maladaptive) left ventricular hypertrophy, finally switching to left ventricular enlargement and progressive heart failure (Iwata 2001 b, Jahns 1999b, Khoynezhad 2007). Both autoantibodies appear to be directed against the second extracellular loop of the respective receptors.
  • myocyte membrane proteins e.g., receptors
  • myocyte membrane proteins e.g., receptors
  • oligopeptides able to form a complex with a MHC or HLA class II molecule of the host.
  • Hoebeke 1996 In case of the human ⁇ 1 -AR computer-based analysis for potential immunogenic amino-acid streches has shown, that the only portion of the receptor molecule containing B- and T-cell epitopes and being accessible to antibodies was in fact the predicted second extracellular receptor loop ⁇ 1 -EC II ) (Hoebeke 1996). This might explain the successful use of second loop-peptides for the generation of ⁇ 1 -specific receptor antibodies in different animal-models (Iwata 2001 b, Jahns.
  • the animals developed progressive left ventricular (LV)-dilatation and dysfunction, a relative decrease in LV wall-thickness, and selective down regutation of ⁇ 1 -AR, a feature that is also seen in human DCM (Lohse 2003).
  • anti- ⁇ 1 -AR induced dilated immune-cardiomyopathy can now be regarded as a pathogenetic disease entity of its own, together with other established receptor-directed autoimmune diseases such as myasthenia gravis or Graves' disease (Freedman 2004, Hershko 2005, Jahns 2004, Jahns 2006).
  • beta-blocking agents in order to attenuate or even abolish the autoantibody-mediated stimulatory effects, at least if ⁇ -blockers can indeed prevent the antibody-induced activation of ⁇ 1 -AR (Freedman 2004, Jahns 2000, Matsui 2001, Jahns 2006).
  • New therapeutic approaches actually include elimination of stimulatory anti- ⁇ 1 -AR by non-selective or selective immunoadsorption (Hershko 2005, Wallukat 2002), or direct targeting of the anti- ⁇ 1 -EC II antibodies and/or the anti- ⁇ 1 -EC II producing B-cells themselves (that is, induction of immune tolerance) (Anderton 2001).
  • Non-selective immunoadsorption because of an increased risk of infection after immunoglobulin depletion, requires the substitution of human IgG on the ground of safety (Felix 2000) with all possible side effects of substituted human proteins known in the art including severe anaphylactic reactions and death.
  • WO 01/21660 discloses certain peptides homologous to epitopes of the 1 st and the 2 nd loop of ⁇ 1 -AR, and proposes to apply these peptides for medical intervention of dilatative cardiomyopathy (DCM). Even if WO 01/21660 mentions marginally that peptides may be modified in order to protect them against serum proteases, for example by cyclization, corresponding examples and embodiments are not given and any in vitro or in vivo effect of the proposed peptides on the course of DCM or on the course of receptor-antibody titers is not shown. Moreover, in WO 01/21660 intends to rely on the above mentioned non-selective immunoadsorption approaches bearing the correspondingly mentioned risks.
  • DCM dilatative cardiomyopathy
  • ⁇ 1 -EC II -homologous cyclopeptides e.g. ⁇ 1 -EC II -CPs
  • ⁇ 1 -EC II -CPs are cyclopeptides containing 3 cysteine residues and hence, can form intramolecular bonds, whereby there is a potential option to form two intramolecular bonds (besides the cyclization between the N- and C-terminus), individually.
  • ⁇ 1 -EC II -CP significantly reduced the amount of circulating anti- ⁇ 1 -EC II antibodies and effectively prevented development of cardiac dilatation and dysfunction (Boivin 2005).
  • the above-mentioned ⁇ 1 -EC II -CPs were also disclosed in WO 2006/103101 .
  • the technical problem underlying the present invention is the provision of improved and easily obtainable means and methods for the medical intervention of diseases related to anti- ⁇ -AR antibodies, particular to anti- ⁇ 1 -EC II antibodies.
  • the present invention relates to ⁇ -AR homologous cyclopeptide-mutants (also termed herein as “cyclic peptides” or “cyclopeptides” and the like), particularly to ⁇ 1 -AR homologous cyclopeptide-mutants, namely ⁇ 1 -EC II homologous cyclopeptide-mutants.
  • cyclic peptides also termed herein as "cyclic peptides” or "cyclopeptides” and the like
  • ⁇ 1 -AR homologous cyclopeptide-mutants namely ⁇ 1 -EC II homologous cyclopeptide-mutants.
  • the structure of these cyclopeptide-mutants/cyclic peptides is characterized by being able to form only one individual intramolecular disulphide bond.
  • the present invention relates to a cyclic peptide of formula I: cyclo(x-x h -Cys-x-x a -X b -x c -x-Cys-y-x i -x) (I), wherein
  • mutant cyclic peptides containing only two cysteines which can form one single defined, individual intramolecular disulfide bond, are also able to inhibit anti- ⁇ -AR antibodies, and are useful in inhibiting stimulatory anti- ⁇ 1 -AR antibodies.
  • inventive cyclic peptides comprising only two cysteines, which can form one single defined, individual intramolecular disulfide bond, can easily be obtained/manufactured, biochemically characterized and purified. This is particularly true when pure fractions of the same cyclopeptide isomers are required.
  • a mixture of cyclopeptide isomers, i.e. stereo-isomers, comprising cyclopeptide isomers with different intramolecular disulphide bonds is avoided.
  • a specific and clean medical product fullfilling GMP standards
  • a Cys ⁇ Ser exchange like that at position 18 of the herein exemplarily and preferably disclosed 25-meric cyclopeptide (formulas VII/IX), at position 17 of the herein exemplarily and preferably disclosed 22-meric cyclopeptide (formula IX') or at position 14 of the herein exemplarily and preferably disclosed 18-meric cyclopeptide (formulas VI/VIII), respectively yields cyclic peptides (Cys-Ser cyclic peptides) with excellent antibody-neutralizing and pharmacological effects in vitro ( Figs.
  • the cardio-protective and immunomodulating activity of the cyclic peptides largely depends on their conformation. It was additionally found out in context of this invention that an introduction of the smallest naturally occuring amino-acid glycine at the (predicted) ring closure site (or at the position corresponding thereto) leads to an enhanced binding of anti- ⁇ 1 -AR autoantibodies, i.e. apparently further enhances the similarity of the 22 AA cyclopeptide with the ECII- ⁇ 1 -AR domain.
  • the appended examples indicate that the cyc22AA cyclopeptides have a significantly higher antibody-blocking efficiency in vivo than other ECII-imitating cyclopeptides larger (i.e., cyc25AA peptides) or smaller (i.e., cyc18AA peptides).
  • Computer-aided modelling studies with said 22 AA cyclopeptide confirmed an excellent imitation of the predicted second extracellular loop structure with a calculated difference in size of only 4.5 Angström (4.5 ⁇ ) at the base of the cyclopeptide (opposed to the assumed antibody-binding site), when compared with the predicted native second extracellular loop backward helix (see also appended Fig. 24 ).
  • the resultant cyclic 22 AA cyclopeptide also represents a biochemically unambiguously defined product (see also Figs. 25 and 26 ).
  • cyclic peptides as disclosed herein show improved features, for example as compared to peptides comprising three Cys residues (for example the Cys/Cys cyclic peptides disclosed in WO 2006/103101 ).
  • improved features of the cyclic peptides of this invention are an extremely good capacity for blocking anti- ⁇ 1 -AR antibodies and their advanced producibility according to GMP standards.
  • the antibody-blocking capacity of mutated cyclopeptides of this invention is not affected by the length of the peptide, as long as the peptide is not shorter than 18 AA and not longer than 25 AA. This was exemplarily demonstrated by the reduction of the number of amino acids of the peptide from 25 to 18. Within the range of 18 to 25 amino acids, cyclic peptides having 22 amino acids are most effective in accordance with this invention and, accordingly, are a particular preferred embodiment. An example of such a particular preferred 22mer cyclic peptide is shown in formula IX'.
  • One advantage of the cyclopeptide mutants of the present invention is -by mutating one particular cysteine (preferably the Cys corresponding to Cys 216 of the amino acid sequence of ⁇ 1 -AR) to a serine-residue and by reinforcing formation of the unique possible intramolecular S-S bridge through a second S-S specific cyclization procedure- that their conformational restraint is increased.
  • this increased restraint of the inventive peptides leads to a molecule that better mimics the epitope presented in the native conformation of the second ⁇ 1 -EC II loop on the cell surface.
  • Beta blockers such as bisoprolol, which are used in the art for the treatment of DCM and other diseases which are caused by stimulatory anti- ⁇ 1 -AR antibodies, significantly reduce both heart rate and blood pressure.
  • an in vivo -application of the mutant cyclopeptides of the present invention has no negative impact on lung function, heart rate or blood pressure ( Figs. 20 and 21 ).
  • a number of important laboratory parameters to assess liver and kidney function were not influenced by the repeated cyclopeptide injections ( Figs. 22a /b and 23). Therefore, the cyclic peptides of the present invention are, inter alia, particularly suitable for the treatment of distinct patient groups which otherwise could not be treated by using a beta blocker, i.e. patients who, for example, already suffer from bradycardia or for whom the use of beta blockers is not possible because of contraindications (like those suffering from obstructive lung disease or hypotension).
  • the cyclic peptides of the present invention can easily be characterized and produced as pure fractions of the same isomer. This leads to a high reproducibility.
  • the particular advantage of the peptides of the present invention is that mixtures of isomers, which have to be separated and must be characterized in laborious testings, are avoided, and that at least one further production step (separation and/or biochemical characterization) is finally omitted (see also Sewald 2002).
  • the present invention is, inter alia, based on the experiments described in the appended examples.
  • one of the cysteines either at position 17 or at position 18 of the ⁇ 1 -EC II 25AA-cyclopeptide was replaced by a serine residue (Cys 17 or 18 ⁇ Ser 17 or 18 mutation), so that only one individual, single intramolecular disulfide bond (S-S) can be formed ( Fig. 1 ).
  • S-S single intramolecular disulfide bond
  • Fig. 1 Measures like this provides the potential to reduce side effects and to maintain or to increase the biological efficacity of the constructs of the present invention.
  • the cyclic peptides of this invention can be obtained, in contrast to the peptides of the prior art which form mixtures of isomers, by simple, robust and highly reproducible manufacturing processes. These can be scaled up efficiently. Furthermore these processes avoids separation of isomers mixtures and are suitable for GMP standards.
  • the appended examples provide for corresponding manufacturing/production methods.
  • the cyclization of the inventive peptides was, inter alia, obtained by the introduction of a "DGlu" mutation, e.g. at the (ring) closure site of the cyclic peptide; Gln ⁇ DGlu mutation as shown in Fig. 2 ).
  • the 18AA cyclopeptide-mutants contained a cysteine ⁇ serine exchange either at position 14 or at position 13 (18AA containing Cys 13 -Ser 14 or Ser 13 -Cys 14 mutant cyclopeptides, respectively), either combined with a (further) glutamine-exchange/D-glutamic acid, e.g. at the ring closure site of the cyclic peptide (Gln ⁇ D-Glu mutation), or not ( Fig. 2 ).
  • the 22AA cyclopeptide-mutants contained a cysteine ⁇ serine exchange at position 17 (22AA containing Cys 16 -Ser 17 ), optionally combined with the introduction of a Gly residue at position 22 (a possibly ring closure site of the cyclic peptide; Fig. 24 ).
  • ⁇ -adrenergic receptors ⁇ -AR
  • ⁇ 1 -AR particulary ⁇ 1 -adrenergic receptors
  • SEQ ID NO. 40 nucleotide and amino acid sequence (SEQ ID NO. 40) of the human ⁇ 1 -AR (also known as adrenergic ⁇ -1-receptor (ADRB1)) can be obtained from databank entry NM_000684 or NP_000675.
  • ⁇ -ARs are known to form two extracellular domains termed herein as EC I and EC II or ⁇ (1) -EC I and ⁇ (1) -EC II .
  • the cyclic peptides of the present invention share sequence similarity with ⁇ 1 -EC II , particularly with the amino acid stretch DEARRCYNDPKCCDFV (SEQ ID NO. 33) or RAESDEARRCYNDPKCCDFVTNR (SEQ ID NO. 34) of the human ⁇ 1 -AR (amino acid positions 204 to 219 or 200 to 222, respectively) or, particularly, with the amino acid stretch DEARRCYNDPK (SEQ ID NO. 45) or ESDEARRCYNDPK (SEQ ID NO. 46) of the human ⁇ 1 -AR.
  • ⁇ -AR as used herein preferably refers to a ⁇ 1 -adrenergic receptor (ß 1 -AR), more preferably to the human ⁇ 1 -AR as described above.
  • a cyclic peptide provided herein has as least one of the features selected from the group consisting of:
  • ⁇ 1 -AR The structure of ⁇ 1 -AR was, inter alia, analyzed by Warne (2008 Nature . DOI:10. 1038).
  • the cyclic peptide of the present invention is defined by the general formula cyclo(x-x h -Cys-x-x a -x b -x c -x-Cys-y-x i -x) (formula I).
  • "y” may be any amino acid residue but Cys, preferably “y” may be any amino acid residue except Pro and exept Cys.
  • "y” may be any amino acid, as long as this amino acid does not form an intramolecular linkage (e.g. a disulphide bond) with another amino acid of the cyclic peptide provided herein (e.g. with another Cys of the cyclic peptide provided herein).
  • "y” may be any amino acid similar to Cys (i.e. having a similar chemical structure and/or a similar behavior within a 3 dimentional peptide structure), with the exeption that it does not form an intramolecular linkage (e.g. a disulphide bond) with another amino acid of the cyclic peptide provided herein (e.g. with another Cys of the cyclic peptide provided herein). More preferably, "y” may be any polar amino acid but Cys, like Thr or Ser. Most preferably, in the cyclic peptide provided herein, "y” is Ser or a Ser analogue.
  • Ser analogue in this context means a residue, particularly an amino acid residue, having a structural character similar to that of Ser.
  • Ser analogue refers to, for example, a(n) (amino acid) residue having a similar chemical structure like that of Ser and/or a similar behavior within a 3 dimentional peptide structure like that of Ser.
  • y may also be selenocysteine or an analogue therof.
  • cyclic peptides of this invention comprise only two Cys able to form an intramolecular linkage.
  • Such cyclic peptides can, for example, be obtained by substituting a third Cys of a peptide homologous to the ⁇ 1 -EC II by a different amino acid.
  • the Cys to be substituted is the one corresponding to the 2 nd or, which is preferred, 3 rd Cys of the ⁇ 1 -EC II which lie in direct proximity to each other (amino acid position 215 and 216 of human ⁇ 1 -AR (see also NP_000675 and SEQ ID NO. 40).
  • Cys residues are referred to herein as "Cys-Cys”, “Cys/Cys”, “Cys 215 -Cys 216 “ or “Cys 215 /Cys 216 " and the like).
  • the resulting mutant peptides or mutations as disclosed herein are accordingly termed as “Cys-Ser”, “Cys/Ser”, “Cys 13 , 16 or 17 -Ser 14, 17 or 18 “ or “Cys 13 , 16 or 17 /Ser 14 , 17 or 18 " mutant peptides or mutations or "Ser-Cys”, “Ser/Cys”, “Ser 13 or 17 -Cys 14 or 18 “ or “Ser 13 or 17/Cys14 or 18 " mutant peptides or mutations, depending which of the Cys is replaced and how many amino acids the mutant peptide comprises.
  • mutant peptides as disclosed herein are defined by referring to the particular amino acid exchanges at a certain position. Then, the mutant peptides/mutations are, for example, termed “Cys 14 , 17 or 18 ⁇ Ser 14 , 17 or 18 " mutant peptides/mutations or "Cys 13 or 17 ⁇ Ser 13 or 17 " mutant peptides/mutations, depending on whether the Cys corresponding to Cys 216 or the Cys corresponding to the Cys 215 , respectively, of ⁇ 1 -AR is replaced by a different amino acid.
  • indices "14, 17 or 18" or “13 or 17” relate to the position in the exemplified cyclic peptide of the invention, whereby position 1 corresponds to the first "x" as defined in formula I, i.e cyclo( x -x h -Cys-x-x a -x b -x c -x-Cys-y-x i -x).
  • indices given above refer to the position of the indicated amino acid within the herein disclosed particular 18mer, 22 mer or 25mer peptide, respectively.
  • the starting point with respect to an indicated amino acid position given for a cyclic peptide disclosed herein is the N-terminal amino acid of the linearized backbone the cyclic peptide (like the first "x" in formula I, see above).
  • the starting point with respect to an indicated amino acid position given for a linear peptide disclosed herein is its N-terminal amino acid.
  • h can be any integer from 1 to 15, preferably from 5 to 9, and/or i can be any integer from 0 to 14, preferably from 1 to 14, more preferably from 0 to 6 and even more preferably from 1 to 6. Accordingly, h can be 1, 2, 3, 4, 5, 5, 6, 7, 8, 9, 10, 11. 12, 13, 14 or 15 and/or i can be 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14. Preferably, h is 5, 8 or 9 and/or i is 3, 4 or 6. More preferably, h is 8 and/or i is 4. In particularly preferred embodiments of this invention, x h stands for the particular amino acid stretches DEARR (SEQ ID NO.
  • x h stands for the particular amino acid stretch AESDEARR (SEQ ID NO. 47) and/or x i stands for the particular amino acid stretch DFVT (SEQ ID NO. 48).
  • the cyclic peptide of the present invention may consist of at least 18 amino acids and of at most 25 amino acids. Accordingly, the cyclic peptide of the present invention may consist of 18, 19, 20, 21, 22, 23, 24 or 25 amino acids, whereby particularly 18 or 25 amino acids are preferred and particularly 22 amino acids are most preferred. In a less preferred embodiment, also smaller peptides, i.e. peptides comprising 16 or 17 amino acids are envisaged.
  • a peptide-length equal or above 26 amino acids may be stimulating directly (that is, without the use of carrier proteins) immunocompetent T-cells and thus may provoke an undesired paradoxal increase of anti- ⁇ 1-receptor antibody production through T-cell mediated B-cell stimulation.
  • a peptide-length below 16 amino acids leads to undesired crystallization during the production process and problems in dissolving the synthesized products in an aqueous solution, e.g. for purposes of i.v. or s.c. injections
  • cyclic peptides falling under the above given definitions a) to f) of formula I and consisting of only 16 amino acids or, even less preferred, consisting of only 17 amino acids are provided.
  • a non-limiting example of such a less preferred cyclic peptide is the peptide cyclo(Ala-Arg-Arg-Cys-Tyr-Asn-Asp-Pro-Lys-Cys-Ser-Asp-Phe-Val-Tyr-Gln/DGlu) (formable by an amino acid backbone as depicted in SEQ ID NO. 39).
  • the disclosed cyclic peptide contains only one Pro. Accordingly, it is particularly preferred that neither the y nor an x of the formulas depicted herein, except of exactly one of x a , x b and x c , is not Pro. Within the amino acid stretch x a , x b and x c as depicted in formula I (or other formulas), it is preferred that x c is Pro.
  • an acidic amino acid like Asp or Glu precedes the Pro contained in the cyclic peptide of the invention.
  • x b as depicted in formula I is an acidic amino acid, like Asp or Glu.
  • x c is Pro
  • x b may be an acidic amino acid
  • x b is Pro
  • x a may be an acidic amino acid
  • the x of formula I (or other formulas) lying between x a and the first Cys may be an acidic amino acid.
  • the cyclic peptide of the present invention may be defined by formula I' or I": cyclo(x I -x h -Cys-x-x a -x b -x c -x-Cys-y-x i -x) (I'); cyclo(X III -x h -Cys-x-x a -x b -x c -x-Cys-y-x i -x) (I'').
  • the cyclic peptide of the present invention may be defined by formula I''' or I"": cyclo(x I -x h -Cys-x-x a -x b -x c -x-Cys-y-x i -x II ) (I'''); cyclo(x III -x h -Cys-x-x a -x b -x c -x-Cys-y-x i -x IV ) (I'''').
  • x I and x II as depicted in formula I' and I''' may, as mentioned, any amino acid but Cys.
  • x I and x II are such amino acids able to form a peptide bond, or the like, with each other under conditions of a "head to tail” cyclization. "Head to tail” cyclizations are known in the art (e.g.
  • x II as referred to in formula I' and I''' can be Gln or Glu, wherein Glu may also be DGlu, (D-Glu; D-Glutamic acid).
  • Glu may also be DGlu, (D-Glu; D-Glutamic acid).
  • X II is Gin.
  • x III and x IV as depicted in formula I''' and I''' may, as mentioned, any amino acid but Cys.
  • x III and x IV are such amino acids able to form a peptide bond, or the like, with each other under conditions of a "head to tail" cyclization.
  • a possible example of an amino acid that may be x III is Arg.
  • One possible, and most preferred, example of an amino acid that may be x IV is Gly or a Gly analogue.
  • Gly analogue in this context means a residue, particularly an amino acid residue, having a structural character similar than that of Gly.
  • Gly analogue refers to, for example, a(n) (amino acid) residue having the same (or even a smaller) size than a Gly residue. It was surprisingly found in context of this invention that particularly a small (amino acid) residue like Gly at the "X IV " position leads to an improved mimicking of the ECII of ⁇ 1-AR by the corresponding cyclic peptides of the invention.
  • the cyclic peptides of this invention lack Trp and/or His. Accordingly, it is particularly envisaged in context of the invention, that neither an x nor y as depicted in any of Formula I to I'''' is Trp or His. Furthermore, it is preferred that the provided cyclic peptides lack sites susceptible for hydrolysis or cleaving proteases, like, for example, serum proteases. The meanings of the terms "hydrolysis” and "(serum) proteases,” are well known in the art.
  • a peptide as provided herein can also be described as a peptide consisting of or comprising an amino acid sequence homologous to SEQ ID NO. 33 (representing a wild type amino acid strech comprising epitopes of ⁇ 1 -EC II ), wherein (a) the amino acid corresponding to position 13 (or, less preferred, corresponding to position 12) of SEQ ID NO. 33 is not Cys and the amino acid corresponding to positions 6 and 12 (or, less preferred, corresponding to position 6 and 13) of SEQ ID NO. 33 is Cys, (b) wherein said amino acid sequence contains no further Cys able to form an intramolecular linkage within the peptide, i.e. within that part of the peptide being homologous to SEQ ID NO.
  • peptide can function as a cyclic peptide in accordance with this invention, e.g. is able to block anti- ⁇ -AR antibodies, or wherein the peptide can form such a cyclic peptide.
  • the further provisions given herein with respect to the structure of the disclosed linear and/or cyclic peptides apply here, mutatis mutandis.
  • the so defined peptide consists of a stretch of 16 amino acids being homologous to SEQ ID NO.
  • amino acids 33 flanked at the N- and C-terminus by one or more amino acids, preferably naturally occurring amino acids, like the "x I “/"x III " at position 1 and the "x II “/”x IV " at the last position of formulas I' to I'''' given herein.
  • homologous means identical on amino acid level for at least 18,75%, at least 37,5%, at least 50%, at least 56,25%, at least 62,5%, at least 68,75%, at least 75%, at least 81,25%, at least 87,5% or 93,75%, wherein the higher values are preferred.
  • amino acid or “amino acid residue” is known in the art and is used herein accordingly. Thereby, it is of note that when an “amino acid” is a component of a peptide/protein the term “amino acid” is used herein in the same sense than "amino acid residue”.
  • an “amino acid” or “amino acid residue” as referred to herein is preferably envisaged to be a naturally occurring amino acid, more preferably a naturally occurring L-amino acid (except the above mentioned DGlu).
  • an “amino acid” or “amino acid residue” in context of this invention may also be a non-naturally occurring (i.e. a synthetic) amino acid, like, for example, norleucine or ß-alanine, or, particularly in case of "y” of the formulas depicted herein, selenocysteine or an analog therof.
  • the term "acidic amino acid(s)” as used herein is intended to mean an amino acid selected from the group comprising Asp, Asn, Glu, and Gln, preferably Asp and Glu;
  • the term "basic amino acid(s)” as used herein is intended to mean an amino acid selected from the group comprising Arg, Lys and His, preferably Arg and Lys;
  • the term “aliphatic amino acid(s)” as used herein is intended to mean any amino acid selected from the group comprising Gly, Ala, Ser, Thr, Val, Leu, Ile, Asp, Asn, Glu, Gin, Arg, Lys, Cys and Met;
  • the term "polar amino acid(s)"as used herein is intended to mean any amino acid selected from the group comprising Cys, Met, Ser, Tyr, Gln, Asn and, less preferred, Trp.
  • the cyclic peptide as provided herein may be a cyclic peptide of formula II, III or III: cyclo(x I -x 1 -x 1 -x-x 2 -x 2 -Cys-x-x a -x b -x c -x-Cys-y-x i -x II ) (II); cyclo(x I -x 2 -x-x 1 -x-x 1 -x 1 -x 1 -x-x 2 -x 2 -Cys-x-x a -x b -x c -x-Cys-y-x i -x II ) (III); cydo (x III,2 -x-x 1 -x-x 1 -x 1 -x-x 2 -x 2 -Cys-x-x a -x b -x c -x-Cys--x-Cys-x
  • the cyclic peptide as provided herein may be a cyclic peptide of formula IV, V or V': cyclo(x I -x 1 -x 1 -x 4 -x 2 -x 2 -Cys-x 3 -x a 5 -x b -x c -x 2 -Cys-y-x 1 -x 3 -x 3 -x II ) (IV); cyclo(x I -x 2 -x 4 -x 1 -x 4 -x 1 -x 1 -x 4 -x 2 -x 2 -Cys-x 3 -x a 5 -x b -x c -x 2 -Cys-y-x 1 -x 3 -x 3 -x 4 -x 5 -x 2 -x II ) (V); cyclo(X III, 2 -x 4 -x 1 -x 4 -x 1 -x 4 -x 2 -
  • the cyclic peptides comprise an amino acid strech as defined by amino acid position 2-12 or 2-14 of formula II or IV, an amino acid strech as defined by amino acid position 4-16 or 4-18 of formula III or V or an amino acid strech as defined by amino acid position 3-15 or 3-17 of formula or III' or V':
  • the cyclic peptide as provided herein may be a cyclic peptide of formula II, III or III'
  • the cyclic peptide as provided herein may comprise the amino acid stretch
  • the cyclic peptide as provided herein may comprise the amino acid stretch
  • the cyclic peptides of this invention comprise one or more epitopes beared by ⁇ 1 -EC II , like, for example, epitopes beared by any of the above mentioned amino acid stretches (or by parts of the disclosed cyclic peptides comprising these amino acid stretches).
  • epitope particularly refers to an amino acid stretch to which an (auto)anti- ⁇ 1 -AR antibody binds.
  • an epitope in context of this invention consists of at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13 or at least 14 amino acids.
  • Non limiting examples of a cyclic peptide according to this invention are cyclic peptides selected from the group consisting of:
  • those cyclic peptides being Cys-Ser mutant peptides, i.e. having the Cys corresponding to the third Cys of the ⁇ 1 -EC II (the Cys at position 216 of ⁇ 1 -AR) exanged by Ser, are particularly preferred.
  • the above given examples refer to such particularly preferred cyclic peptides.
  • such cyclic peptides are particularly useful in inhibiting or diagnosing anti- ⁇ 1 -AR antibodies.
  • Non limiting examples of less preferred cyclic peptides according to this invention are cyclic peptides selected from the group consisting of:
  • cyclic peptides according to this invention are particularly those cyclic peptides, the pharmacological and/or diagnostic function of which has been demonstrated in the appended examples (e.g. those characterized by any one of formula VI to IX').
  • the skilled person is, one the one hand, readily in the position to find out corresponding variants of the peptides of the invention.
  • the skilled person is able to test whether a given variant of peptides of the present invention still has the desired function, for example the ability to specifically bind to ⁇ -AR antibodies, and therefore has the potential for a corresponding medical intervention, like the therapeutic and diagnostic applications described and provided herein.
  • Corresponding experimental guidance for such tests, i.e. respective assays are exemplarily provided and described herein, particularly in the appended examples.
  • variants of the herein disclosed and described peptides are still functionally active in accordance with this invention, i. e. functionally active as binding partners for anti- ⁇ -AR antibodies, particularly for anti- ⁇ 1 -AR antibodies against the ⁇ 1 -EC II , more particularly functionally active as inhibitors of ⁇ 1 -AR and even more preferably active in inhibiting the interaction between ⁇ 1 -AR and anti- ⁇ 1 -AR antibodies against the ⁇ 1 -EC II , more preferably auto-anti- ⁇ 1 -AR antibodies against the ⁇ 1 -EC II ; and, second, that these variants are not present in form of isomers mixtures or do not form isomers mixtures when cyclized in accordance with production method of this invention.
  • These variants are envisaged to have only two certain Cys residues forming or being able to form only one individual intramolecular linkage (e.g. disulphide bond).
  • amino acids of said peptides of the present invention are replaced by other one or more naturally occurring or synthetic amino acids.
  • this/these amino acid exchange(s) is/are (a)conservative amino acid exchange(s), i.e. that the replacement amino acid belongs to the same category of amino acids than the amino acid to be replaced.
  • an acidic amino acid may be replaced by another acidic amino acid
  • a basic amino acid may be replaced by another basic amino acid
  • an aliphatic amino acid may be replaced by another aliphatic amino acid
  • a polar amino acid may be replaced by another polar amino acid.
  • variants of the (cyclo) peptides of the present invention are variants wherein at least one of an acidic amino acid of is replaced by a different amino acid selected from the group consisting of acidic amino acids, at least one of the basic amino acids is replaced by a different amino acid selected from the group consisting of basic amino acids, at least one of a polar amino acid is replaced by a different amino acid selected from the group consisting of polar amino acids and/or at least one of an aliphatic amino acid is replaced by a different amino acid selected from the group consisting of aliphatic amino acids (given that the above mentioned-requirements are fulfilled).
  • amino acid exchanges which lead to variants of the disclosed (cyclic) peptides are such, that the pattern of polarity and charge within the tertiary structure of the resulting variant still substantially mimics the three-dimensional structure of the corresponding EC II epitope(s) of ⁇ 1 -AR.
  • the herein defined Cys may also be replaced by other amino acids, as long as the replacement still leads to an individual intramolecular linkage, like that of a disulphide bond, within the cyclopeptide, i.e. the avoidance of isomers mixtures formation during cyclization and/or a correct mimicry of the EC II of ⁇ 1 -AR.
  • Such amino acid may, inter alia, be a non-naturally occurring amino acid, like a non-naturally occurring amino acid having an -SH group able to form a disulphide bond.
  • the Cys given in formula I, above is indeed a naturally occurring amino acid, preferably Cys itself.
  • amino acids forming the (cyclic) peptide of the present invention may be modified.
  • any amino acid as used herein may also represent its modified form.
  • an alanine residue as used herein may comprise a modified alanine residue.
  • Such modifications may, among others, be a methylation or acylation or the like, whereby such modification or modified amino acid is preferably comprised by the present invention as long as the thus modified amino acid and more particularly the peptide containing said thus modified amino acid is still functionally active as defined herein, like functionally active as an inhibitor of anti- ⁇ 1 -AR antibodies, preferably active in inhibiting the interaction between ⁇ 1 -AR and antibodies, and more preferably auto-antibodies directed against ⁇ 1 -AR.
  • Respective assays for determining whether such a peptide, i. e. a peptide comprising one or several modified amino acids, fulfils this requirement are known to the one skilled in the art and, among others, also described herein, particularly in the example part hereof.
  • the invention also provides derivatives of the disclosed (cyclic) peptides such as salts with physiologic organic and anorganic acids like HCl, H 2 SO 4 , H 3 PO 4 , malic acid, fumaric acid, citronic acid, tatratic acid, acetic acid.
  • sequences of the peptides disclosed are indicated from the N-terminus to the C-terminus, whereby the N-terminus is at the left side and the C-terminus is at the right side of the respective depicted amino acid sequence.
  • the corresponding sequences are indicated from the side corresponding to the left side of formula I to the side corresponding to the right side of formula I.
  • a "cyclic peptide” or “cyclopeptide” and the like in accordance with the present invention is a peptide intramolecularly forming a molecular ring structure within its amino acid backbone/primary amino acid sequence by at least one, preferably by at least two, more preferably by exactly two intramolecular linkages having covalent character.
  • the forming of this molecular ring structure is, in context of this invention, also termed "cyclization".
  • the cyclic peptide of this invention has two intramolecular linkages having covalent character, wherein one of these linkages is an intramolecular linkage between the N- and C-terminal ends of a peptide being the amino acid backbone/primary amino acid sequence of the cyclic peptide disclosed and the other one is an intramolecular linkage between two non-terminal amino acids of this peptide.
  • these two non terminal amino acids may be two Cys.
  • the peptide bond as mentioned throughout this invention can be formed by the NH 2 group of an N-terminal amino acid and the COOH group of an C-terminal amino acid of a peptide forming the amino acid backbone/primary amino acid sequence of the cyclic peptide disclosed.
  • an intramolecular linkage between the N- and C-terminal ends of a peptide forming the amino acid backbone/primary amino acid sequence of the cyclic peptide disclosed is a peptide bond and an intramolecular linkage between two non-terminal amino acids of this peptide is an S-S linkage (i. e. disulphide bond).
  • an intramolecular S-S linkage within the cyclic peptide provided can be formed between two Cys residues within the amino acid backbone/primary amino acid sequence of said cyclic peptide.
  • cyclic peptides of this invention not only the above mentioned two particular intramolecular covalent linkage may be formed but also further intramolecular linkages may occur, with the proviso that the herein described functionality of the cyclic peptides is maintained and that the cyclic peptides can still easily be characterized biochemically, which, e.g., means that no isomers mixtures are formed during cyclization of the corresponding amino acid backbone/primary amino acid sequence.
  • Such further intramolecular linkages are additional bonds formed by a side chain of NH 2 groups and COOH groups of the constituent amino acids.
  • amino acid backbone or “primary amino acid sequence” as used throughout the present invention refer, on the one hand, to that structural component or part of a cyclic peptide which is formable or formed by its corresponding amino acid sequence. On the other hand, these terms refer to the linear peptides able to form the cyclic peptides of this invention by cyclization.
  • a cyclic peptide which is obtainable by the method for producing a cyclic peptide as provided herein.
  • the definitions given herein-above also apply with respect to this particularly provided cyclic peptide of the present invention.
  • the disclosed cyclic peptides are formable by or are formed by.
  • these peptides are the linear peptides forming or being able to form the herein disclosed cyclic peptides, i.e. the amino acid backbone/primary amino acid sequence thereof.
  • such a linear peptide can be any peptide, the covalent linkage of the N- and C-terminus of which results in a cyclic peptide as disclosed in accordance with the present invention.
  • such a linear peptide may be some kind of an intermediate compound in an procedure of producing the cyclic peptides of this invention, like the method for producing a cyclic peptide as disclosed herein.
  • N- and C-terminal end of a linear peptide provided herein may be any amino acid pair lying in direct proximity to each other within the amino acid backbone of a cyclic peptide disclosed in context of this invention.
  • cyclization (ring closure) of the cyclic peptide of this invention may generally occur between any of said amino acid pairs.
  • the skilled person is readily in the position to find out such particular amino acid pairs which are effective/suitable to act as N- and C-terminal ends of a herein disclosed linear peptide, i.e. which are effective/suitable to act as an amino acid pair being involved in the ring closure/cyclization as defined in context of this invention.
  • the cyclization (ring closure) of a linear peptide of this invention may occur between Ala and Gln or Glu, i.e. the N-terminal amino acid of this linear peptide would be Ala and the C-terminal amino acid would be Gln or Glu.
  • linear peptides able to form the cyclic peptide of the present invention are SEQ ID NO. 1 to 4 and, less preferred SEQ ID NO. 5 to 8.
  • the cyclization (ring closure) of a linear peptide of this invention may occur between Lys and Pro, i.e. the N-terminal amino acid of this linear peptide would be Lys and the C-terminal amino acid would be Pro.
  • Examples of such linear peptides able to form the cyclic peptide of the present invention are SEQ ID NO. 17 to 20 and, less preferred SEQ ID NO. 21 to 24.
  • the cyclization (ring closure) of a linear peptide of this invention may occur between Arg and Gly, i.e. the N-terminal amino acid of this linear peptide would be Arg and the C-terminal amino acid would be Gly.
  • An examples of such a linear peptide able to form the cyclic peptide of the present invention is SEQ ID NO. 41.
  • the cyclization (ring closure) of a linear peptide of this invention may occur between Lys and Pro, i.e. the N-terminal amino acid of this linear peptide would be Lys and the C-terminal amino acid would be Pro.
  • An examples of such a linear peptide able to form the cyclic peptide of the present invention is SEQ ID NO. 43.
  • the cyclic peptides of the invention may further comprise (e.g. have covalently bound) (a) further substituent(s), like labels, anchors (like proteinaceous membrane anchors), tags (like HIS tags) and the like.
  • substituents and methods for adding them to the cyclic peptided of this invention are known to those of ordinary skill in the art.
  • labels in this context include, inter alia, fluorochromes (like fluorescein, rhodamine, Texas Red, etc.), enzymes (like horse radish peroxidase, ⁇ -galactosidase, alkaline phosphatase), radioactive isotopes (like 32P, 33P, 35S, 125I or 1231, 135I, 124I, 11C, 150), biotin, digoxygenin, colloidal metals, chemi- or bioluminescent compounds (like dioxetanes, luminol or acridiniums).
  • fluorochromes like fluorescein, rhodamine, Texas Red, etc.
  • enzymes like horse radish peroxidase, ⁇ -galactosidase, alkaline phosphatase
  • radioactive isotopes like 32P, 33P, 35S, 125I or 1231, 135I, 124I, 11C, 150
  • One particularly envisaged label that may be bound to the peptide of this invention is a fluorochrome belonging to a FRET pair of fluorochromes, for example a GFP variant (e.g. GFP, eGFP, EYFP or ECFP).
  • a GFP variant e.g. GFP, eGFP, EYFP or ECFP.
  • biomolecules A variety of techniques are available for labeling biomolecules, are well known to the person skilled in the art and are considered to be within the scope of the present invention and comprise, inter alia, covalent coupling of enzymes or biotinyl groups, phosphorylations, biotinyiations, random priming, nick-translations, tailing (using terminal transferases).
  • Detection methods comprise, but are not limited to, autoradiography, fluorescence microscopy, direct and indirect enzymatic reactions, etc.
  • the substituent(s) can be bound (e.g. covalently) to the cyclic peptides of the invention directly or via linkers.
  • linkers The skilled person is readily in the position to find out appropriate linkers to be employed in this context.
  • the present invention relates to a nucleic acid molecule comprising a nucleotide sequence encoding the amino acid backbone/primary amino acid sequence of a cyclic peptide as disclosed in context of this invention.
  • the present invention also relates to a nucleic acid molecule comprising a nucleotide sequence encoding the linear peptides as provided and described herein.
  • such nucleic acid molecule may comprise a nucleotide sequence as depicted in any one of SEQ ID NO. 42, 44, 9 to 12, 25 to 28, 49, 50, 53 and 54 or, less preferred, SEQ ID NO. 13 to 16 and 29 to 32 or a nucleotide sequence which differs therefrom due to the degeneracy of the genetic code.
  • nucleic acid molecule(s) As well known in the art and are used accordingly in context of the present invention.
  • nucleotide sequences and/or nucleic acid sequences/molecules refer to all forms of naturally occurring or recombinantly generated types of nucleotide sequences and/or nucleic acid sequences/molecules as well as to chemically synthesized nucleotide sequences and/or nucleic acid sequences/molecules.
  • nucleic acid analogues and nucleic acid derivatives such as e. g. locked DNA, PNA, oligonucleotide thiophosphates and substituted ribo-oligonucleotides.
  • these terms also refer to any molecule that comprises nucleotides or Nucleotide analogues.
  • nucleic acid molecule(s) refers to deoxyribonucleic acid (DNA) or ribonucleic acid (RNA).
  • the "nucleic acid molecule(s)”, “nucleic acid sequence(s)” and “nucleotide sequence(s)” may be made by synthetic chemical methodology known to one of ordinary skill in the art, or by the use of recombinant technology, or may be isolated from natural sources, or by a combination thereof.
  • the DNA and RNA may optionally comprise unnatural nucleotides and may be single or double stranded.
  • Nucleic acid molecule(s) also refer to sense and anti-sense DNA and RNA, that is, a nucleotide sequence which is complementary to a specific sequence of nucleotides in DNA and/or RNA.
  • nucleic acid molecule(s) may refer to DNA or RNA or hybrids thereof or any modification thereof that is known in the state of the art (see, e.g., US 5525711 , US 4711955 , US 5792608 or EP 302175 for examples of modifications).
  • These molecules of the invention may be single- or double-stranded, linear or circular, natural or synthetic, and without any size limitation.
  • nucleic acid molecule(s) may be genomic DNA, cDNA, mRNA, antisense RNA, ribozymal or a DNA encoding such RNAs or chimeroplasts ( Cole-Strauss Science 1996 273(5280) 1386-9 ). They may be in the form of a plasmid or of viral DNA or RNA.
  • Nucleic acid molecule(s) may also refer to (an) oligonucleotide(s), wherein any of the state of the art modifications such as phosphothioates or peptide nucleic acids (PNA) are included.
  • PNA peptide nucleic acids
  • nucleic acid molecules as provided herein are particularly useful for producing a cyclic peptide of the invention, for example by the corresponding method disclosed herein.
  • the present invention also relates to a vector comprising the nucleic acid molecule as disclosed herein and described above.
  • Said vector may be a cloning vector or an expression vector, for example, a phage, plasmid, viral or retroviral vector.
  • Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host/cells.
  • the herein provided nucleic acid molecule may be joined to a particular vector containing selectable markers for propagation in a host.
  • a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate or rubidium chloride precipitate, or in a complex with a charged lipid or in carbon-based clusters, such as fullerens. Should the vector be a virus, it may be packaged in vitro using an appropriate packaging cell line prior to application to host cells.
  • the nucleic acid molecule of this invention is operatively linked to expression control sequences (e.g. within the herein disclosed vector) allowing expression in prokaryotic or eukaryotic cells or isolated fractions thereof.
  • Expression of said polynucleotide comprises transcription of the nucleic acid molecule, preferably into a translatable mRNA.
  • Regulatory elements ensuring expression in eukaryotic cells preferably mammalian cells, are well known to those skilled in the art. They usually comprise regulatory sequences ensuring initiation of transcription and optionally poly-A signals ensuring termination of transcription and stabilization of the transcript. Additional regulatory elements may include transcriptional as well as translational enhancers.
  • Possible regulatory elements permitting expression in prokaryotic host cells comprise, e.g., the lac, trp or tac promoter in E. coli, and examples for regulatory elements permitting expression in eukaryotic host cells are the AOX1 or GAL1 promoter in yeast or the CMV-, SV40-, RSV-promoter (Rous sarcoma virus), CMV-enhancer, SV40-enhancer or a globin intron in mammalian and other animal cells.
  • Beside elements which are responsible for the initiation of transcription such regulatory elements may also comprise transcription termination signals, such as the SV40-poly-A site or the tk-poly-A site, downstream of the polynucleotide.
  • suitable expression vectors are known in the art such as Okayama-Berg cDNA expression vector pcDV1 (Pharmacia), pCDM8, pRc/CMV, pcDNA1, pcDNA3 (Invitrogen), pSPORT1 (GIBCO BRL).
  • said vector is an expression vector and/or a gene transfer vector.
  • Expression vectors derived from viruses such as retroviruses, adenoviruses, vaccinia virus, adeno-associated virus, herpes viruses, or bovine papilloma virus, may be used for delivery of the polynucleotides or vector of the invention into a targeted cell population.
  • isolated fractions thereof refers to fractions of eukaryotic or prokaryotic cells or tissues which are capable of transcribing or transcribing and translating RNA from the vector of the invention.
  • Said fractions comprise proteins which are required for transcription of RNA or transcription of RNA and translation of said RNA into a polypeptide.
  • Said isolated fractions may be, e.g., nuclear and cytoplasmic fractions of eukaryotic cells such as of reticulocytes.
  • Kits for transcribing and translating RNA which encompass the said isolated fractions of cells or tissues are commercially available, e.g., as TNT reticulolysate (Promega).
  • vectors as provided and described herein are particularly useful for producing a cyclic peptide of the invention, for example by the corresponding method disclosed herein.
  • the present invention relates to a recombinant host cell comprising the nucleic acid molecule and/or the vector as disclosed herein.
  • the nucleic acid molecule and/or the vector as disclosed herein can, inter alia, be used for genetically engineering host cells, e.g., in order to express and isolate the amino acid backbone/primary amino acid sequence of the cyclic peptide disclosed herein, and hence, the linear peptide of this invention.
  • Said host cell may be a prokaryotic or eukaryotic cell; see supra.
  • the nucleic acid molecule or vector which is present in the host cell may either be integrated into the genome of the host cell or it may be maintained extrachromosomally.
  • the host cell can be any prokaryotic or eukaryotic cell, such as a bacterial, insect, fungal, plant, animal, mammalian or, preferably, human cell.
  • Preferred fungal cells are, for example, those of the genus Saccharomyces, in particular those of the species S. cerevisiae, or those belonging to the group of hyphal fungi, for example several penicillia or aspergilla strains.
  • prokaryotic is meant to include all bacteria which can be transformed or transfected with a nucleic acid molecule for the expression of an amino acid backbone/primary amino acid sequence of the cyclic peptide disclosed herein, and hence, the linear peptide of this invention.
  • Prokaryotic hosts may include gram negative as well as gram positive bacteria such as, for example, E. coli, S. typhimurium, Serratia marcescens and Bacillus subtilis.
  • a nucleic acid molecule coding for an amino acid backbone/primary amino acid sequence of the cyclic peptide disclosed herein, and hence, the linear peptide of this invention, can be used to transform or transfect a host using any of the techniques commonly known to those of ordinary skill in the art. Methods for preparing fused, operably linked genes and expressing them in bacteria or animal cells are well-known in the art (Sambrook, supra). The genetic constructs and methods described therein can be utilized for expression of the above mentioned amino acid backbone/primary amino acid sequence and linear peptide in, e.g., prokaryotic hosts.
  • expression vectors containing promoter sequences which facilitate the efficient transcription of the inserted polynucleotide are used in connection with the host.
  • the expression vector typically contains an origin of replication, a promoter, and a terminator, as well as specific genes which are capable of providing phenotypic selection of the transformed cells.
  • the transformed prokaryotic hosts can be grown in fermentors and cultured according to techniques known in the art to achieve optimal cell growth.
  • the expressed peptides can then be isolated from the grown medium, cellular lysates, or cellular membrane fractions.
  • the isolation and purification of the microbially or otherwise expressed peptides may be by any conventional means such as, for example, preparative chromatographic separations and immunological separations such as those involving the use of monoclonal or polyclonal antibodies ( Ausubel, Current Protocols in Molecular Biology, Green Publishing Associates and Wiley Interscience, N.Y. (1994 )).
  • the corresponding host cells are particularly useful for producing a cyclic peptide of the invention, for example by the corresponding method disclosed herein.
  • the present invention relates to a method for producing a cyclic peptide of the present invention, comprising the steps of
  • cyclization encompasses both, forming of the intramolecular bridge (disulphide bond) and the ring closure by covalently connecting the N- and C-termini of the backbones of the cyclic peptides to be produced.
  • the N-terminal amino acid of the amino acid backbone/linear peptide to be cyclized in order to produce a cyclic peptide of this invention is Ala, Arg or Lys and the corresponding C-terminal amino acid is Gln, Gly or Glu (also DGlu is possible) or Pro.
  • N- and C-terminal amino acids are envisaged, i.e. also other cyclization (ring closure) sites can be employed in context of the disclosed method.
  • the cyclopeptide mutants were first synthesized in form of their linear peptides/amino acid backbones (for example by applying a chemical synthesis approach, like the Fmoc / tert butyl strategy (as described in WO 2006/103101 ; Chen W.C. and White P.D.: Fmoc Solid Phase Peptide Synthesis, Oxford University Press 2003 )), and were then cyclized covalently on the backbone by condensation of the C-terminal carboxyl group with the amino group of the N-terminal amino acid ("head to tail" cyclization; Kates S. and Albericio F.: Solid phase synthesis, CRC-Press, 2000 ).
  • a disulphide bond is established between those two cysteine residues of the linear peptides which are able to form a disulphide bond (e.g. between Cys 7 and Cys 13 of the 18mer peptide, between Cys 10 and Cys 16 of the 22mer peptide or between Cys 11 and Cys 17 of the 25mer peptide) by chemical interaction known in the art (e.g. Benoiton N.L.: Chemistry of Petide Synthesis. CRC-Press, 2005 ).
  • the ring closure of the linear backbone of the cyclic peptides to be produced may be performed before or after the formation of the S-S bridge.
  • the S-S bridge between the two Cys residues of the AA chain of the peptides may be the first step in the "cyclization" procedure of the described production process and the ring closure may be the second step, or vice versa.
  • the skilled person is able to find out which of these particular approaches is appropriate for a given setup of the production preconditions.
  • linear peptides/amino acid backbones of the cyclic peptides to be produced can also be produced by recombinant engineering techniques. Such techniques are well known in the art (e. g. Sambrook, supra). As also mentioned above, by this kind of production of said linear peptides/amino acid backbones particular advantage can be taken of the herein disclosed and described nucleic acid molecules, vectors and/or host cells. The definitions correspondingly given above apply here, mutatis mutandis.
  • this invention also relates to a cyclic peptide obtainable or obtained by the above described method, but also to a corresponding linear peptide (amino acid backbone/ primary sequence of the corresponding cyclic peptide) obtainable or obtained by the above described method as some kind of an intermediate product (particularly a product obtainable or obtained by step a) of the above described method).
  • the cyclic peptide according to the present invention may, inter alia, be used in medical intervention approaches.
  • Such approaches comprise the use as or in a diagnostic agent and for the manufacture of a medicament for the treatment of diseases or the use in or as a composition, preferably a pharmaceutical composition, a diagnostic composition or a diagnostic kit, preferably for the detection of anti- ⁇ -AR antibodies, more preferably for the detection of anti- ⁇ 1 -AR antibodies.
  • the antibodies as defined or described herein are preferably autoantibodies.
  • Non-limiting uses and applications of the compounds, particularly the cyclic peptide according to the present invention are described herein, for example in the following.
  • the present invention also relates to a composition
  • a composition comprising a cyclic or a linear peptide, a nucleic acid molecule, a vector or a recombinant host cell as disclosed and provided in context of the present invention, and optionally a carrier.
  • said composition is a pharmaceutical composition and said carrier is a pharmaceutically acceptable carrier.
  • composition of this invention is particularly useful when employed in the treatment, amelioration or prevention as described and defined herein. Accordingly, the pharmaceutical composition of this invention may be used for the treatment, amelioration or prevention of a disease where the activity of a ⁇ -AR is enhanced or for the treatment of a patient having antibodies against a ⁇ -AR. Moreover, the pharmaceutical composition of this invention may be used for inducing immune tolerance of a patient, particularly immune tolerance of a patient with respect to immunogenic stretches of the endogenous ⁇ 1 -AR.
  • the (pharmaceutical) composition provided may either comprise two or a plurality (like at least 3 or at least 5) of cyclic peptides of the present invention.
  • cyclic peptides may be administered to a patient in need of medical intervention in accordance with the present invention.
  • administration of said more than one of cyclic peptides may be simultaneously or successively.
  • the present invention relates to the pharmaceutical composition, the method or uses for medical intervention or the cyclic peptide or the pharmaceutical composition as disclosed herein, wherein said cyclic peptide is administered with or said pharmaceutical composition comprises at least one further pharmaceutically active agent.
  • Said at least one further pharmaceutically active agent may be a ⁇ receptor blocker, preferably a selective ⁇ -AR blocker, like, for example, a ⁇ 1 -AR blocker selected from the group consisting of atenolol, metoprolol, nebivolol, bisoprolol and the like.
  • a selective ⁇ -AR blocker like, for example, a ⁇ 1 -AR blocker selected from the group consisting of atenolol, metoprolol, nebivolol, bisoprolol and the like.
  • this kind of particular combination provides for protection from antibody-induced, selective ⁇ 1 -AR downregulation by the herein provided cyclic peptides, since ⁇ 1 -AR is at the same time upregulated by betablockers, like bisoprolol or metoprolol, and ultimately results in a synergistic effect of the cyclic peptides and the additional ⁇ -blocker(s).
  • the carrier optionally comprised in the (pharmaceutical) composition of the invention or to be administered together with the (pharmaceutical) composition or the cyclic peptide of the invention may particularly be a pharmaceutically acceptable carrier, excipient or diluent.
  • Such carriers are well known in the art. The skilled person is readily in the position to find out such carriers which are particularly suitable to be employed in accordance with the present invention.
  • compounds of the invention may be formulated in aqueous solution, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiologically saline buffer.
  • physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiologically saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • compositions of the present invention in particular those formulated as solutions, may be administered parenterally, such as by intravenous injection.
  • pharmaceutically acceptable carriers well known in the art into dosages suitable for subcutaneous or oral administration.
  • Such carriers enable the compounds according to the present invention to be formulated as tablets, pills, capsules, dragees, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a subject to be treated.
  • Liposomes are spherical lipid bilayers with aqueous interiors. All molecules present in an aqueous solution at the time of liposome formation are incorporated into the aqueous interior. The liposomal contents are both protected from the external microenvironment and, because liposomes fuse with cell membranes, are efficiently delivered near the cell surface. Delivery systems involving liposomes are disclosed in U.S. Patent No. 4,880,635 to Janoff et al . The publications and patents provide useful descriptions of techniques for liposome drug delivery.
  • compositions comprising a compound according to the present invention for parenteral and/or subcutaneous administration include aqueous solutions of the active
  • suspensions of the active compounds may be prepared as appropriate oily injection suspensions.
  • lipophilic solvents or vehicles include fatty oils such as sesame oil or castor oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injections suspensions may contain compounds which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, dextran, or the like.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions and to allow for a constantly slow release of the substance in the organism.
  • the disclosed pharmaceutical composition or cyclic peptide may be administered in a pharmaceutically/therapeutically effective dose, which means that a pharmaceutically/therapeutically effective amount of the compound administered is reached.
  • a pharmaceutically/therapeutically effective dose refers to that amount of the compound administered (active ingredient) that produces amelioration of symptoms or a prolongatian of survival of a subject which can be determined by the one skilled in the art doing routine testing.
  • the dosage regimen of the compounds to be administered in accordance with the present invention will be determined by the attending physician and clinical factors. As is well known in the medical arts, that dosages for any one patient depends upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently. A person skilled in the art is aware of and is able to test the relevant doses, the compounds to be medically applied in accordance with the present invention are to be administered in.
  • the effect of the cyclic peptides provided herein namely the blockage of anti- ⁇ 1 -AR antibodies, can be obtained in a dose dependent manner.
  • the efficiency of the Cys ⁇ Ser mutated cyclopeptides as disclosed herein depends on a threshold concentration ( Figs. 14 , 15 , and 16B,C ).
  • the disclosed pharmaceutical composition or cyclic peptide may particularly be administered in a manner that it is present in a concentration, i.e. reaches a threshold concentration, of at least 0,05 mg per kg body weight, preferably in a concentration of at least 0,1 mg per kg body weight, more preferably in a range of 0,1 mg per kg body weight (100 ⁇ g/kg) to 100 mg per kg body weight, more preferably in a range of 1 mg per kg body weight to 100 mg per kg body weight and most preferably in a range of 1 mg per kg body weight to 10 mg per kg body weight.
  • the effective dose of the disclosed pharmaceutical composition or cyclic peptide may be at about 1 mg per kg body weight.
  • higher concentrations of the disclosed pharmaceutical composition or cyclic peptide are generally envisaged to be reached by correspondingly applied administration schemes.
  • such higher concentrations may be at least 2, 3, 4 or 5 mg per kg body weight. Concentrations of at least 1 mg per kg body weight or at least 2 mg per kg body weight are preferred.
  • One particularly preferred, non-limiting administration scheme to be applied in context of this invention is an s.c. or i.v. application every two or four weeks.
  • a dose of 1 to 4 mg/kg i.v. every other month were sufficient to obtain therapeutic levels of the compounds according to the present invention, with the respective dosage for humans preferably being about 0,3-10 mg/kg i.v. or s.c, more preferably being about 1-10 mg/kg i.v. or s.c., even more preferably being about 1-5 mg/kg i.v. or s.c.
  • the administration of the disclosed cyclic peptides may initially trigger a transient opposite immune response, in particular w/hen applied in higher doses.
  • transient immune responses in the lomg run are compensated by the antibody-inactivating activity of the administered cyclic peptides.
  • This may lead to an decelerated effect of the administered cyclic peptides, i.e. a decelerated elimination of anti- ⁇ 1 -AR antibodies and hence a decelerated reduction of (aberrant) ⁇ 1 -AR activity.
  • the present invention also relates to a method for
  • the present invention also relates to a cyclic peptide or a pharmaceutical composition as disclosed herein, and optionally a pharmaceutically acceptable carrier, for
  • the diseases to be medically intervened (treated, ameliorated, prevented or diagnosed) in accordance with this invention or the diseases the patient as defined and described herein suffers from are preferably those, where the ⁇ 1 -AR is activated in a non-physiological manner, more preferably is activated by antibodies, more preferably by auto-antibodies which are directed against the ⁇ 1 -AR.
  • the diseases to be medically intervened in accordance with this invention or the diseases the patient as defined and described herein suffers from comprise, however, are not limited thereto, the group of heart diseases.
  • the heart diseases to be medically intervened in accordance with this invention or the heart diseases the patient as defined and described herein suffers from may comprise but are not limited to infectious and non-infectious heart disease, ischemic and non-ischemic heart disease, inflammatory heart disease and myocarditis, cardiac dilatation, idiopathic cardiomyopathy, (idiopathic) dilated cardiomyopathy (DCM), immune-cardiomyopathy, heart failure, and any cardiac arrhythmia including ventricular and/or supraventricular premature capture beats as well as any atrial arrhythmia including atrial fibrillation and/or atrial flutter.
  • infectious and non-infectious heart disease ischemic and non-ischemic heart disease
  • inflammatory heart disease and myocarditis cardiac dilatation
  • idiopathic cardiomyopathy idiopathic dilated cardiomyopathy (DCM)
  • DCM dilated cardiomyopathy
  • immune-cardiomyopathy heart failure
  • the heart disease as referred to in the descriptions and definitions given herein with respect to the methods or the cyclic peptide or the pharmaceutical composition of the invention may be heart diseases selected from the group comprising infectious and non-infectious heart disease, ischemic and non-ischemic heart disease, inflammatory heart disease and myocarditis, cardiac dilatation, idiopathic cardio-myopathy, (idiopathic) dilated cardiomyopathy (DCM), immune-cardiomyopathy, heart failure, and any cardiac arrhythmia including ventricular and/or supraventricular premature capture beats as well as any atrial arrhythmia including atrial fibrillation and/or atrial flutter.
  • heart diseases selected from the group comprising infectious and non-infectious heart disease, ischemic and non-ischemic heart disease, inflammatory heart disease and myocarditis, cardiac dilatation, idiopathic cardio-myopathy, (idiopathic) dilated cardiomyopathy (DCM), immune-cardiomyopathy, heart failure, and any cardiac arrhythmi
  • DCM preferably idiopathic DCM.
  • a particular subgroup of the "patients" for the purpose of the present invention are those patients suffering from any of the diseases described herein, more particularly the group of heart diseases described herein and having at the same time antibodies directed against ⁇ -ARs, more preferably antibodies against the ⁇ 1 -AR, whereby the antibodies are preferably auto-antibodies.
  • a disease to be medically intervened (treated, ameliorated, prevented or diagnosed) in accordance with this invention or a disease the patient as defined and described herein suffers from is intended to be induced by antibodies against a ⁇ -AR, preferably by antibodies against ⁇ 1 -AR.
  • these antibodies are auto-antibodies.
  • the means and methods provided herein are particularly useful when provided in the prophylaxis/prevention of a disease as defined herein.
  • a patient may be treated with the cyclic peptide and/or pharmaceutical composition of the invention prior to the onset (of symptoms) of a disease as defined herein.
  • this preventive treatment may follow a preceding diagnostic application that, e.g., takes advantage of the diagnostic means and methods provided herein.
  • a preventive treatment taking advantage of the therapeutic means and methods of this invention is applied, when the risk to develop a disease as defined herein is diagnosed, e.g. when anti- ⁇ -AR (auto-)antibodies are detected.
  • a preferred "patient” is one bearing at risk to develop a disease as defined herein.
  • a patient is one having anti- ⁇ -AR (auto-)antibodies, preferably anti- ⁇ 1 -AR (auto-)antibodies, but not (yet) suffering from a disease as defined herein, or symptoms thereof.
  • the immune tolerance to be induced in context of this invention is envisaged to be particularly obtained by suppression of the production of antibodies against immunogenic stretches of the ⁇ -AR molecule, which, without being bound by theory, may be due to a blockade of the antigen-recognition sites of the antibody-producing early (mature) B-cells and memory B-cells.
  • the provided pharmaceutical composition or cyclic peptide is particularly useful for the treatment, prevention and/or amelioration of any of the diseases and patient groups or patients as defined herein including the detection of anti- ⁇ -AR antibodies in these patients by using the aforementioned compounds.
  • a "patient” for the purposes of the present invention i. e. to whom a compound according to the present invention is to be administered or who suffers from the disease as defined and described herein or who is intended to be diagnosed in accordance with this invention, includes both humans and other animals and organisms.
  • the compounds and methods of this invention are applicable to or in connection with both human therapy and veterinary applications including diagnostic(s), diagnostic procedures and methods as well as staging procedures and methods.
  • the patient is a mammal, and in the most preferred embodiment the patient is human.
  • mutant cyclic peptides according to the present invention may also be used for the preparation of a medicament for the treatment, prevention and/or amelioration of any of the diseases and patient groups/patients as defined herein.
  • What is said herein for the pharmaceutical composition applies also to the medicament for the manufacture of which the peptides of the present invention may be used.
  • the present invention is related to a diagnostic agent comprising or being a cyclic peptide or a composition according to this invention, and optionally at least one further biologically active compound.
  • the herein disclosed diagnostic agent consists of or comprises a mutant peptide of the present invention, whereby the mutant peptide comprises a label.
  • a label may be selected from the group comprising radioactive labels and fluorescent labels.
  • Respective labels are known to the ones skilled in the art. The definitions and descriptions of labels as given herein-above apply here, muatis mutandis.
  • the peptide is the part of the diagnostic agent conferring specific binding characteristics to the diagnostic agent, preferably binding to anti- ⁇ 1 -AR antibodies, whereas the label confers the signalling characteristics to the diagnostic agent.
  • the diagnostic agent of this invention may comprise, apart from (a) labelled or unlabelled mutant peptide(s) of the present invention, a further biologically active compound.
  • a further biologically active compound may be a means to confer signalling characteristics to the diagnostic agent, particularly in case the mutant peptides of the present invention are unlabelled.
  • the further biologically active compound can be an antibody, preferably a monoclonal antibody, and more preferably a labelled antibody specifically binding to a mutant peptide of the present invention or to a complex consisting of a mutant peptide of the present invention and an anti- ⁇ -AR antibody, preferably an anti- ⁇ 1 -AR antibody.
  • the present invention relates to a method for diagnosing a disease as defined and described herein comprising the steps of
  • the present invention is related to a method for diagnosing a patient which can be treated using the mutant peptides, pharmaceutical compositions and medicaments according to the present invention.
  • a step of detecting antibodies against a ⁇ -AR (for example in a sample) using the compounds of the present invention and/or a step of considering whether the outcome of said detection step indicates a disease as defined herein may be employed.
  • a disease as defined herein or the risk to develop a disease as defined herein is indicated, when the titer of said anti- ⁇ -AR antibodies is increased.
  • the present invention relates to a cyclic peptide, a composition or a diagnostic agent as provided and described herein for diagnosing (for example in a sample) a disease as defined herein.
  • a disease as defined herein or the risk to develop a disease as defined herein is indicated by an increased titer of anti- ⁇ -AR antibodies.
  • the term "increased titer of anti- ⁇ -AR antibodies” means that the titer of anti- ⁇ -AR antibodies (for example in a sample derived from a patient to be diagnosed in accordance with this invention) is higher than that of a healthy control patient, i.e. a patient not suffering from a disease as defined herein and/or a patient lacking anti- ⁇ -AR antibodies.
  • an "increased titer of anti- ⁇ -AR antibodies" in accordance with the present invention preferably refers to any occurrence of anti- ⁇ -AR antibodies, i.e. any occurrence of a detectable amount of anti- ⁇ -AR antibodies.
  • sample in accordance with the present invention includes, but is not limited to, (a) biological or medical sample(s), like, e.g. (a) sample(s) comprising cell(s) or tissue(s).
  • sample(s) may comprise(s) biological material of biopsies.
  • biopsies comprise cell(s) or tissue(s) taken, e. g. by the attending physician, from a patient/subject as described herein.
  • the biological or medical sample to be analysed in context of the present invention is or is derived from blood, plasma, white blood cells, urine, semen, sputum, cerebrospinal fluid, lymph or lymphatic tissues or cells, muscle cells, heart cells, cells from veins or arteries, nerve cells, cells from spinal cord, brain cells, liver cells, kidney cells, cells from the intestinal tract, cells from the testis, cells from the urogenital tract, colon cells, skin, bone, bone marrow, placenta, amniotic fluid, hair, hair and/or follicles, stem cells (embryonic, neuronal, and/or others) or primary or immortalized cell lines (lymphocytes, macrophages, or cell lines).
  • samples in accordance with the present invention are those derived from blood or plasma.
  • the biological or medical sample as defined herein may also be or be derived from biopsies, for example biopsies derived from heart tissue, veins or arteries.
  • the present invention relates to a diagnostic kit, for example a diagnostic kit for the detection of antibodies against a ⁇ -AR, comprising the cyclic peptide, composition or diagnostic agent of the invention.
  • the kit in accordance with the present invention comprises at least one of the compounds as disclosed according to the invention, like, for example a cyclic or linear peptide of the present invention, a nucleic acid molecule, vector or host cell of the invention or a composition or diagnostic agent according to the present invention.
  • the kit further comprises an instruction leaflet, and/or a buffer for use in the application of the kit, and/or at least one reaction vessel for carrying out the detection reaction for which the kit is or is to be used.
  • at least one, some or all of the reagents used in connection with the application of said kit are present as portions useful in carrying out the reaction(s) for which the kit is to be used.
  • the cyclic peptides, diagnostic agents or kits of this invention may also be applied for the detection of ⁇ 1 -AR. Accordingly, the cyclic peptides, diagnostic agents or kits of this invention may particularly be useful for identifying/detecting bound anti- ⁇ 1 -AR antibodies on cell- and/or tissue surfaces.
  • the cyclic peptides, diagnostic agents or kits of this invention may be used for in imaging purposes, like single photone emission computed tomography (SPECT), MiBi, PET, magnetic resonance tomography (MRT) or other diagnostic imaging techniques employed in medicine. Due to the 131l-labelled CP-distribution pattern in vivo ( Fig. 30 ), the cyclic peptides, diagnostic agents of this invention may, for example, be particularly useful as organ-specific tracers.
  • One particular approach for using the compounds according to the present invention as a diagnostic and in a diagnostic method, respectively, is a three-step screening procedure.
  • this method comprises performing an ELISA with the cyclic peptides according to the present invention as well as determining immunofluorescence and determining cAMP responses in cells expressing native human ⁇ -AR. It is to be acknowledged that each and any of the aforementioned steps can as such be preformed for the detection of said antibodies using the cyclic peptides according to the present invention.
  • a large number of patients, for example heart failure patients, may thus be screened for functionally active anti- ⁇ 1 -AR antibodies.
  • functionary active anti- ⁇ 1 -AR antibodies is preferably based on their effects on receptor-mediated signalling, that is, their effects on cellular cAMP levels and on the activity of the cAMP-dependent protein kinase (PKA).
  • Cyclic AMP is an universal second messenger of many G protein-coupled receptors including the ⁇ -AR family. It exerts its effects via PKA, cAMP-gated ion channels, phosphodiesterases, and exchange proteins directly activated by cAMP, known as Epac1 and 2.
  • Epac1 and 2 exchange proteins directly activated by cAMP
  • Fluorescence resonance energy transfer between green fluorescent protein (GFP) variants fused to the regulatory and catalytic subunits of PKA has been described to study the spatio-temporal dynamics of cAMP in neurons ( Hempel CM, Vincent P, Adams SR, Tsien RY, Selverston Al. Nature. 1996;384:113-114 ) or cardiac myocytes. ( Zaccolo M, Pozzan T., Science. 2002:295:1711-1715 ).
  • single chain fluorescence indicators have been described in the art which are characterized by having an enhanced cyan (CFP) or yellow fluorescent protein (YFP) directly fused to the cAMP-binding domain of Epac-proteins, which allowed to achieve a higher sensitivity and better temporal resolution of the cAMP measurements.
  • CFP enhanced cyan
  • YFP yellow fluorescent protein
  • Such system is, among others described in W02005/052186 .
  • Such system can be used in connection with any diagnostic procedure using the cyclic peptides or other corresponding compounds according to the present invention.
  • Also such system can be used for, however is not limited thereto, analyzing the prevalence of functionally active anti- ⁇ 1 -AR antibodies.
  • Such diagnostic method is applied to a cohort of previously antibody-typed DCM patients or any individual to be assessed insofar or any individual suspected of suffering from any of the diseases described herein or being at risk to suffer therefrom.
  • the ability of ⁇ -blockers to inhibit anti- ⁇ 1 -AR antibodies-induced receptor activation may be assessed and determined, respectively.
  • the afore described assay which is a FRET-based method as described in WO 2005/052186 making use of the peptides according to the present invention is advantageous insofar as it is simpler, less time consuming, and at the same time discloses or identifies all DCM patients previously considered anti- ⁇ 1 -EC II antibody-positive.
  • This embodiment of a FRET based method of diagnosing making use of one or several of the peptides according to the present invention is based on detecting antibody-induced increases in cAMP.
  • the present invention is also related to the use of one or several of the peptides according to the present invention for use in an Epac-FRET assay. More preferably such Epac-FRET assay is used for diagnosis, even more preferably for the diagnosis of patients suffering from or suspected of suffering from any of the disease described herein.
  • the present invention relates to a method for detecting of antibodies against a ⁇ -AR (for example in a sample as defined herein) comprising the step of contacting the cyclic peptide of the invention with said antibodies to be detected.
  • the present invention relates to the cyclic peptide, composition or diagnostic agent as disclosed herein for detecting (for example in a sample as defined herein) antibodies against a ⁇ -AR.
  • ab antibody
  • Abs or abs antibodies
  • AR adrenergic receptor
  • EC extra cellular domain of a ⁇ -AR EC II extra cellular domain II of a ⁇ -AR and AA amino acid.
  • cyclopeptides which can form only one single individual disulfide bond are composed of 18, 22 or 25 amino acids (AA): EC II -18AA Cys/Ser mutant (Gln-)cyclopeptide, EC II -22AA Cys/Ser mutant (Gly-)cyclopeptide and EC II -25AA Cys/Ser mutant (Gln-)cyclopeptide, respectively.
  • the primary sequence is partially homologous to the human sequence of the ⁇ 1 -AR (amino acid positions 204 through 219, 200 through 220 and 200 through 222, respectively).
  • the 18AA, 22 or 25AA cyclopeptide mutant adopts a conformation which more closely mimics that of the epitope as presented on the surface of the native ⁇ 1 -EC II protein loop.
  • cyclization has often been employed as a tool to prolong the duration of action of peptide, since in general cyclic peptides are more stable to proteolysis than their linear counterparts.
  • Cyclization can occur between Cys 7 and Cys 13 (disulphide bond) and Ala 1 and Gln 18 (ring closure).
  • Cyclization can occur between Cys 10 and Cys 16 (disulphide bond) and Arg 1 and Gly 22 (ring closure).
  • Cyclization can occur between Cys 11 and Cys 17 (disulphide bond) and Ala 1 and Gln 25 (ring closure).
  • the cyclopeptide mutants of the present invention are first synthesized as linear peptides, and are then cyclized covalently on the backbone by condensation of the C-terminal carboxyl group with the amino group of the N-terminal amino acid. Subsequently, a disulphide bond between cysteine residues 7 and 13 (18mer cyclopeptide), cysteine residues 10 and 16 (22mer cyclopeptide) and cysteine residues 11 and 17 (25mer cyclopeptide) is established.
  • the linear peptide is assembled by stepwise solid phase peptide synthesis using an Fmoc/tert butyl strategy. Chlorotrityl is used as a starting resin.
  • the first amino acid (Fmoc-Pro-OH) is coupling with DIEA in DMF, the second with PYBOP/ HOBT/ DIEA in DMF and the following amino acids with diisopropylcarboimide, HOBT in DMF.
  • the peptide quality is monitored online by UV detection.
  • Deprotection / coupling (two-fold excess) procedure is described below: Table 1: Deprotection / coupling (two-fold excess) procedure Step Solvents Cycle 1 Coupling / DMF (*) min Coupling 2 DMF 3 x 1 min Wash 3 Piperidine 25% / DMF 1 min Deprotection 4 Piperidine 25% / DMF 2 x 15 min Deprotections 5 DMF 7 x 1 min Wash * coupling time is determined by Kaiser test
  • the fully protected peptide with reactive N-terminal amino- and C-terminal carboxylgroups is cleaved from the resin by treatment with hexafluoroisopropanol/ dichloromethane.
  • the "head to tail"-cyclization of the protected peptide is performed with PyBOP/ NaHCO 3 in high DMF dilution (10 mmol of linear peptide / 1 L of DMF). The cyclization is completed after 3 days. After DMF evaporation, the peptide is washed with 5% NaHCO 3 , H 2 O and pure H 2 O. The reaction mixture is cooled down, and the peptide is deprotected excepting the cysteine groups. Afterwards, the partially protected peptide is isolated by precipitation with methyl t-butyl ether.
  • the crude peptide is pre-purified by liquid chromatography: Stationary phase: silica C18, 15 ⁇ m, 120 A Eluant: H 2 O acetonitrile + 0.1% TFA Detection: UV (210 nm)
  • the disulfide cyclization is performed in H 2 O (2 mg/mL) with the presence of dimethyl sulfoxyde (3%). The cyclization reaction is completed after 3 days.
  • the peptide is purified by HPLC, using the conditions described above.
  • the fractions with a purity greater than 95% are pooled.
  • the peptide is exchanged on an ion exchange resin (Dowex 1X2) and the final solution lyophilized.
  • the peptide content is determined by amino acid analysis (Edman sequencing).
  • the blocking capacity of 18AA Cys/Ser mutants was highest (69 ⁇ 2%), followed by 25AA Cys/Cys (67 ⁇ 2%), 18AA Cys/Cys cyclopeptides (60 ⁇ 4%), and 3 Cys-containing linear 25AA peptides (55 ⁇ 4%) compared with the totally inefficient cyclic 25AA or 18AA Ser/Cys mutants (6 ⁇ 2% or 1 ⁇ 2%, respectively; P ⁇ 5x 10 -30 or P ⁇ 3.7x10 -33 ).
  • Ser/Cys mutants had almost no inhibitory effect, irrespective of the amino-acid at the closure site (6 ⁇ 2%, 25AA Ser/Cys, P ⁇ 5x 10 -30 ; 1 ⁇ 2%, 18AA Ser/Cys with Gln- (P ⁇ 3.7x19 -33 ) or 1 ⁇ 2% with D-Glu-closure, ( P ⁇ 6x10 -55 ).
  • Linear peptides were clearly less efficient, yielding a dose-dependent decrease in receptor antibody-titers of only 24% [8-fold excess], 35% [20-fold excess], and about 50% at a 80-fold excess for both, linear 25AA Cys/Cys and linear 18AA Cys/Ser peptides.
  • the normalized YFP/CFP-ratio of the registered FRET emission signals served to quantify the effect of the cyclopetide mutants of the present invention in terms of blockade (in per cent) of antibody-induced cellular cAMP-production of transiently Epac1-transfected stably ⁇ 1 -AR expressing human embryonic kidney cells (HEK 293- ⁇ 1 cells).
  • the x-axis in Fig. 6 corresponds to the registration time given in seconds (s).
  • the inhibitory effect of ⁇ 1 -EC II -18AA cyclopeptide mutants on the antibody-induced stimulation of ⁇ 1 -adrenergic transmembrane signalling was analyzed using an approach by fluorescence resonance energy transfer (FRET). Again, also in terms of inhibiting measurable functional antibody-effects (blocking intracellular cAMP-increases) the cyclic ⁇ 1 -EC II -18AA Cys/Ser mutant was largely superior to its Ser/Cys counterpart, and even slightly more effective than a 3 Cys-containing 25AA Cys/Cys cyclopeptide ( Fig. 6 ).
  • results of the tests performed herein demonstrate that the antibody-blocking capacity of mutated cyclopeptides was not affected by the reduction of the number of amino-acids from a 25-meric to a 18-meric peptide.
  • results also demonstrate an excellent comparability of 25AA Cys/Cys and 18AA Cys/Cys cyclopeptides with the cyclic 25AA or 18AA Cys/Ser mutants, but not with the cyclic 25AA or 18AA Ser/Cys mutants.
  • the animal model used in this example and any other example described herein, if not indicated to the contrary, is the human analogue rat model.
  • this human analogue rat model was treated as described herein-below using the various compounds of the present invention, more particularly compounds of formula VI, VII, VIII and IX, and, as controls, a linear EC II -18AA Cys/Ser mutated (Gln 18 -)peptide (with the following amino-acid sequence: Ala-Asp-Glu-Ala-Arg-Arg-Cys-Tyr-Asn-Asp-Pro-Lys-Cys-Ser-Asp-Phe-Val-Gln) and a linear non-mutated 3 Cys-containing EC II -25AA Cys/Cys (Gln 25 ) peptide (with the following amino-acid sequence: Ala-Arg-Ala-Glu-Ser-Asp-Glu-Ala-Arg-Arg-Cy
  • ASC specific anti-beta1-ECII antibody-secreting cells
  • the in vivo blocking effects of therapeutically used 25AA and 18AA Cys/Ser cyclo-peptide mutants, the 18AA Ser/Cys mutant (Gln-)cyclopeptide, and a mutated linear 18AA Cys/Ser peptide were assessed after a first intravenous (i.v.) dose (i.e., 1.0 mg/kg body weight (Bw)) of each construct injected into long term immunized anti-beta1-ECII antibody-positive rats, yet presenting a cardiomyopathic phenotype (after nine months of 1x monthly immuni-zation with the beta1-ECII/GST antigen; Figs. 14-18 and 17 ).
  • i.v. 1.0 mg/kg body weight
  • Sera were drawn 18-20 hours after the first i.v. injection of the various constructions and assayed for reactivity by ELISA using the 3 Cys-containing linear 25AA Cys/Cys-peptide as an antigen.
  • "Therapeutic" application of various cyclopeptides in cardiomyopathic antibody-positive rats revealed a higher in vivo blocking capacity of 18AA Cys/Ser cyclopeptide mutants (1mg/kg/Bw) compared with either 25AA Cys/Ser cyclopeptide mutants or the clearly less efficient 18AA Ser/Cys cyclopeptide mutants at a same concentration ( Figs. 14 and 15 ).
  • the animals were immunized with a fusion protein containing bacterial glutathione-S-transferase and the sequence of the second extra-cellular loop of the human ⁇ 1 -adrenergic receptor (GST/ ⁇ 1 -EC II ).
  • GST/ ⁇ 1 -EC II human ⁇ 1 -adrenergic receptor
  • progressive dilated immune cardiomyopathy is observed after 6 to 8 months of regular immunization every four weeks ( Fig. 17 ). All of the immunized animals developed high titers of stimulatory anti- ⁇ 1 -EC II antibodies.
  • the specific anti- ⁇ 1 -EC II titer reached a maximum between 6 and 8 months of continuously boosting the animals every 4 weeks, whereas NaCl-injected control animals developed no specific receptor antibodies ( Fig. 18 ).
  • Such immunized animals were used for the application of mutant cyclopeptides according to the present invention.
  • the in-vivo blocking effect of both 25AA and 18AA Cys/Ser mutant cyclopeptides were determined after a first intravenous (i.v.) injection of 1.0 mg/kg body weight (Bw) (for 18AA Cys/Ser cyclopeptides also 0.25 mg/kg/Bw) into immunized antibody-positive rats.
  • Sera were drawn 18-20 hours after i.v. injection of the different peptides and assayed for reactivity by ELISA using the 3 Cys-containing linear 25AA Cys/Cys-peptide as an antigen.
  • the in vivo data confirmed a higher blocking capacity of the 18AA Cys/Ser mutated cyclo-peptides (1mg/kg/Bw) compared with either 25AA Cys/Ser mutants or the clearly less effective 18AA Ser/Cys mutated cyclopeptides at a same concentration ( Figs. 14 and 15 ), The in vivo efficiency of the 18AA Cys/Ser cyclopeptide was also largely superior to that of the linear 18AA Cys/Ser peptide.
  • echocardiographic follow-up data after 4 months of treatment also show a superiority of cyc22AA mutants compared with the cyc18AA Cys/Ser mutant regarding their cardioprotective effects in vivo, as assessed by the decrease in both left ventricular end-diastolic (LVED) and end-systolic (LVES) diameters ( Fig. 29A ), and an increase in "Cardiac Index" (CI, given in ml/min/g body weight; see Fig. 29B ), as determined by 2 dimensional- and Doppler-echocardiography using a Visual Sonics echocardiographic system (Vevo 770, version V2.2.3), equipped with a 17.5 MHz transducer).
  • LVED left ventricular end-diastolic
  • LVES end-systolic
  • an intramolecularly localized disulfide bridge is essential to stabilize and maintain the three-dimensional structure of the construction.
  • the remaining cysteines i.e. in position 209 and 215, in case 216 has been mutated to Ser
  • maintain a defined intramolecular distance further strengthened by introduction of the smallest naturally occuring amino-acid glycine at the (predicted) ring closure site, in order to allow for the formation of a structure-defining intramolecular disulfide bridge.
  • the present invention refers to the following nucleotide and amino acid sequences:
  • SER stands for any nucleotide triplet coding for Ser (serine), i.e. for tcn or agy
  • GLY stands for any nucleotide triplet coding for Gly (Glycine), i.e. for ggn.
  • n stands for any nucleotide, particularly a, c, g or t, y stands for t or c and r stands for a or g.
  • sequences of the various peptides are indicated from the N-terminus to the C-terminus, whereby the N-terminus is at the left side and the C-terminus is at the right side of the respective depicted amino acid sequence.
  • amino acid 3-letter code: 1-letter code: Alanine Ala A Arginine Arg R Asparagine Asn N Aspartic acid Asp D Cysteine Cys C Glutamic acid Glu E Glutamine Gln Q Glycine Gly G Histidine His H Isoleucine Ile I Leucine Leu L Lysine Lys K Methionine Met M Phenylalanine Phe F Proline Pro P Serine Ser S Threonine Thr T Tryptophan Trp W Tyrosine Tyr Y Valine Val V Asparagine or aspartic acid Asx B Glutamine or glutamic acid Glx Z Leucine or Isoleucine Xle J Unspecified or unknown amino acid Xaa X
  • the present invention relates to the following items:

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Cardiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Genetics & Genomics (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Vascular Medicine (AREA)
  • Hospice & Palliative Care (AREA)
  • Transplantation (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Peptides Or Proteins (AREA)
EP12178671.9A 2007-08-24 2008-08-22 Mutante doppelt zyklierte Rezeptorpeptide zur Hemmung von beta-1-Adrenozeptorantikörper Active EP2559698B1 (de)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP12178671.9A EP2559698B1 (de) 2007-08-24 2008-08-22 Mutante doppelt zyklierte Rezeptorpeptide zur Hemmung von beta-1-Adrenozeptorantikörper

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP07016637 2007-08-24
EP08801676A EP2197900B9 (de) 2007-08-24 2008-08-22 Beta 1-adrenozeptor-antikörper inhibierende, mutierte, doppelt cyclisierte rezeptorpeptide
EP12178671.9A EP2559698B1 (de) 2007-08-24 2008-08-22 Mutante doppelt zyklierte Rezeptorpeptide zur Hemmung von beta-1-Adrenozeptorantikörper

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
EP08801676.1 Division 2008-08-22
EP08801676A Division EP2197900B9 (de) 2007-08-24 2008-08-22 Beta 1-adrenozeptor-antikörper inhibierende, mutierte, doppelt cyclisierte rezeptorpeptide

Publications (3)

Publication Number Publication Date
EP2559698A2 true EP2559698A2 (de) 2013-02-20
EP2559698A3 EP2559698A3 (de) 2013-03-06
EP2559698B1 EP2559698B1 (de) 2014-11-12

Family

ID=40130559

Family Applications (3)

Application Number Title Priority Date Filing Date
EP12170992A Withdrawn EP2497777A3 (de) 2007-08-24 2008-08-22 Beta 1-Adrenozeptor-Antikörper inhibierende, mutierte, doppelt cyclisierte Rezeptorpeptide
EP12178671.9A Active EP2559698B1 (de) 2007-08-24 2008-08-22 Mutante doppelt zyklierte Rezeptorpeptide zur Hemmung von beta-1-Adrenozeptorantikörper
EP08801676A Active EP2197900B9 (de) 2007-08-24 2008-08-22 Beta 1-adrenozeptor-antikörper inhibierende, mutierte, doppelt cyclisierte rezeptorpeptide

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP12170992A Withdrawn EP2497777A3 (de) 2007-08-24 2008-08-22 Beta 1-Adrenozeptor-Antikörper inhibierende, mutierte, doppelt cyclisierte Rezeptorpeptide

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP08801676A Active EP2197900B9 (de) 2007-08-24 2008-08-22 Beta 1-adrenozeptor-antikörper inhibierende, mutierte, doppelt cyclisierte rezeptorpeptide

Country Status (14)

Country Link
US (1) US9062095B2 (de)
EP (3) EP2497777A3 (de)
JP (3) JP5248610B2 (de)
CN (2) CN103992406A (de)
AU (1) AU2008291296B2 (de)
CA (1) CA2697108C (de)
DK (1) DK2197900T3 (de)
ES (1) ES2391358T3 (de)
HK (1) HK1176363A1 (de)
HR (1) HRP20120827T1 (de)
PL (1) PL2197900T3 (de)
PT (1) PT2197900E (de)
SI (1) SI2197900T1 (de)
WO (1) WO2009027063A2 (de)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL2197900T3 (pl) * 2007-08-24 2012-12-31 Univ Wuerzburg J Maximilians Zmutowane podwójnie cyklizowane peptydy receptorowe hamujące przeciwciała przeciwko beta 1-adrenoreceptorowi
BRPI1007007A2 (pt) * 2009-01-27 2016-03-22 Julius Maximilians Universität Würzburg peptídeos cíclico, molécula de ácido nucleico, vetor, célula hospedeira, método para produção de um peptídeo cíclico, composição farmacêutica, método para detecção de anticorpos contra um b-ar (em uma amostra)
DE102010018878B4 (de) 2009-04-30 2013-09-26 Julius-Maximilians-Universität Würzburg Neue Zell-Linie zur Fluoreszenz-basierten Detektion von funktionell aktiven Antikörpern und Autoantikörpern gegen den Beta1-adrenergen Rezeptor
EP2402016A1 (de) * 2010-06-29 2012-01-04 Charité - Universitätsmedizin Berlin Aptamere, die eine Interaktion zwischen einem Antikörper und dem ersten oder zweiten extrazellulären loop des menschlichen Beta-Adrenergenrezeptors hemmen
US20140273015A1 (en) 2011-06-10 2014-09-18 Corim- Mun Gmbh BINDING COMPOUNDS TO HUMAN Beta 1-ADRENORECEPTOR (Beta 1-AR) AND THEIR USE IN MEASUREMENT OF AUTO-ANTI- Beta 1-AR ANTIBODIES
AU2013221317B2 (en) 2012-02-16 2017-07-06 Pangu Biopharma Limited Histidyl-tRNA synthetases for treating autoimmune and inflammatory diseases
DK3460054T3 (da) 2013-03-15 2021-01-18 Atyr Pharma Inc Histidyl-tRNA-syntetase-Fc-konjugater
WO2015022953A1 (ja) * 2013-08-14 2015-02-19 株式会社新日本科学 血液脳関門障害改善剤
AU2018256435A1 (en) 2017-04-20 2019-11-07 Atyr Pharma, Inc. Compositions and methods for treating lung inflammation
CN108218963B (zh) * 2018-01-19 2020-10-13 首都医科大学 一种环肽及其制备方法和用途
MA55296A (fr) * 2019-03-14 2022-03-23 Hoffmann La Roche Traitement du cancer avec des anticorps bispécifiques de her2xcd3 en combinaison avec un mab anti-her2
US11986536B2 (en) 2019-03-23 2024-05-21 Ablevia Biotech Gmbh Compound for the sequestration of undesirable antibodies in a patient
EP3715374A1 (de) 2019-03-23 2020-09-30 Ablevia biotech GmbH Verbindung zur sequestrierung von unerwünschten antikörpern in einem patienten
EP4217402A1 (de) * 2020-09-23 2023-08-02 Ablevia biotech GmbH Verbindung zur prävention oder behandlung von autoantikörpervermittelten erkrankungen
CN112266330B (zh) * 2020-10-15 2023-02-24 长沙博源医疗科技有限公司 一种甲氧基肾上腺素衍生物、免疫原、抗甲氧基肾上腺素特异性抗体及其制备方法与应用
WO2023028590A1 (en) * 2021-08-27 2023-03-02 Yale University Molecular degraders of extracellular proteins
JP2023117892A (ja) * 2022-02-14 2023-08-24 公益財団法人 鷹揚郷 ヘプシジン結合ペプチド

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4711955A (en) 1981-04-17 1987-12-08 Yale University Modified nucleotides and methods of preparing and using same
EP0302175A2 (de) 1982-06-23 1989-02-08 Enzo Biochem, Inc. Markierte modifizierte Nukleotide und Polynukleotide und Verfahren zu deren Herstellung, Verwendung und Detektion
US4880635A (en) 1984-08-08 1989-11-14 The Liposome Company, Inc. Dehydrated liposomes
US5525711A (en) 1994-05-18 1996-06-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pteridine nucleotide analogs as fluorescent DNA probes
US5792608A (en) 1991-12-12 1998-08-11 Gilead Sciences, Inc. Nuclease stable and binding competent oligomers and methods for their use
WO2001021660A1 (de) 1999-09-21 2001-03-29 Affina Immuntechnik Gmbh Peptide gegen dcm hervorrufende autoantikörper
WO2005052186A1 (en) 2003-11-26 2005-06-09 Bayerische Julius-Maximilians-Universität Würzburg Means and methods for the determination of camp in vitro and in vivo
WO2006103101A2 (en) 2005-03-31 2006-10-05 Julius-Maximilians-Universität Würzburg MEANS FOR THE INHIBITION OF ANTI-β1-ADRENERGIC RECEPTOR ANTIBODIES

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1433325A (zh) * 1999-11-05 2003-07-30 加利福尼亚大学董事会 通过体内基因递送治疗心血管疾病的技术和组合物
US20040001827A1 (en) * 2002-06-28 2004-01-01 Dennis Mark S. Serum albumin binding peptides for tumor targeting
JP4541490B2 (ja) * 2000-04-07 2010-09-08 株式会社カネカ 拡張型心筋症用吸着体
WO2001083693A2 (en) * 2000-04-28 2001-11-08 Glaxo Group Limited Compounds having affinity for the vascular endothelial growth factor receptor-2 (vegfr-2) and associated uses
CN1988914A (zh) * 2001-08-23 2007-06-27 西兰制药公司 胞间通讯易化化合物的新医学用途
US7041790B2 (en) * 2002-03-26 2006-05-09 Dyax Corp. Fibrinogen binding moieties
US20080206264A1 (en) * 2003-02-04 2008-08-28 New York University Constrained Hiv V3 Loop Peptides as Novel Immunogens and Receptor Antagonists
PL2197900T3 (pl) * 2007-08-24 2012-12-31 Univ Wuerzburg J Maximilians Zmutowane podwójnie cyklizowane peptydy receptorowe hamujące przeciwciała przeciwko beta 1-adrenoreceptorowi

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4711955A (en) 1981-04-17 1987-12-08 Yale University Modified nucleotides and methods of preparing and using same
EP0302175A2 (de) 1982-06-23 1989-02-08 Enzo Biochem, Inc. Markierte modifizierte Nukleotide und Polynukleotide und Verfahren zu deren Herstellung, Verwendung und Detektion
US4880635A (en) 1984-08-08 1989-11-14 The Liposome Company, Inc. Dehydrated liposomes
US4880635B1 (en) 1984-08-08 1996-07-02 Liposome Company Dehydrated liposomes
US5792608A (en) 1991-12-12 1998-08-11 Gilead Sciences, Inc. Nuclease stable and binding competent oligomers and methods for their use
US5525711A (en) 1994-05-18 1996-06-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pteridine nucleotide analogs as fluorescent DNA probes
WO2001021660A1 (de) 1999-09-21 2001-03-29 Affina Immuntechnik Gmbh Peptide gegen dcm hervorrufende autoantikörper
WO2005052186A1 (en) 2003-11-26 2005-06-09 Bayerische Julius-Maximilians-Universität Würzburg Means and methods for the determination of camp in vitro and in vivo
WO2006103101A2 (en) 2005-03-31 2006-10-05 Julius-Maximilians-Universität Würzburg MEANS FOR THE INHIBITION OF ANTI-β1-ADRENERGIC RECEPTOR ANTIBODIES

Non-Patent Citations (83)

* Cited by examiner, † Cited by third party
Title
"Circulation", vol. 115, 2007, AMERICAN, HEART, ASSOCIATION, article "Heart disease and stroke statistics", pages: E69 - E171
"Immunochemical methods in cell and molecular biology", 1987, ACADEMIC PRESS
"Methods in Enzymology", ACADEMIC PRESS, INC.
ANDERTON, IMMUNOLOGY, vol. 104, 2001, pages 367 - 376
AUSUBEL: "Current Protocols in Molecular Biology", 1994, GREEN PUBLISHING ASSOCIATES AND WILEY INTERSCIENCE
BABA, EUR. HEART J., vol. 25, 2004, pages 1108 - 1115
BENOITON N.L.: "Chemistry of Petide Synthesis", 2005, CRC-PRESS
BENOITON: "Chemistry of Peptide Synthesis", 2005, CRC-PRESS
BENOITON: "Chemistry of Peptide Synthesis.", 2005, CRC-PRESS
BOIVIN, EUR. J. HEART FAIL. 4, vol. 24, no. 1, 2005, pages 104
CAFORIO, EUR. J. HEART FAIL., vol. 4, 2002, pages 411 - 417
CHEN W.C.; WHITE P.D.: "Fmoc Solid Phase Peptide Synthesis", 2003, OXFORD UNIVERSITY PRESS
CHIALE, CIRCULATION, vol. 103, 2001, pages 1765 - 1771
CHIEN, ONCOL., vol. 27, 2000, pages 9 - 17
CHRIST, J. MOL. CELL CARDIOL., vol. 41, 2006, pages 716 - 723
CHRIST, J. MOL. CELL. CARDIOL., vol. 33, 2001, pages 1515 - 1525
COLE-STRAUSS, SCIENCE, vol. 273, no. 5280, 1996, pages 1386 - 9
DAVIS LG; DIBMER MD: "Basic methods in molecular biology", 1990, ELSEVIER
ELIES, J. IMMUNOL., vol. 157, 1996, pages 4203 - 4211
ENGELHARDT, PROC. NATL. ACAD. SCI. USA, vol. 96, 1999, pages 7059 - 7064
ERIKSSON, NAT. MED., vol. 9, 2003, pages 1484 - 1490
FABRIZIO, DRUGS THER., vol. 8, 1994, pages 89 - 94
FELIX, J. AM. COLL. CARDIOL., vol. 35, 2000, pages 1590 - 1598
FERRARI, J. EXP. MED., vol. 182, 1995, pages 59 - 65
FREEDMAN, J. CLIN. INVEST., vol. 113, 2004, pages 1379 - 1382
FRIELLE ET AL., PNAS, vol. 84, 1987, pages 7920 - 7924
FRIELLE, PNAS, vol. 84, 1987, pages 7920 - 7924
FU, J. CIIN. INVEST., vol. 91, 1993, pages 1964 - 1968
G6SER, CIRCULATION, vol. 114, 2006, pages 1693 - 1702
HEMPEL CM; VINCENT P; ADAMS SR; TSIEN RY; SELVERSTON AL, NATURE, vol. 384, 1996, pages 113 - 114
HERSHKO, ANN. N.Y. ACAD. SCI., vol. 1051, 2005, pages 635 - 646
HOEBEKE, LNT. J. CARDIOL., vol. 54, 1996, pages 103 - 111
JAHNS, CIRCULATION, vol. 99, 1999, pages 649 - 654
JAHNS, EUR. J. PHARMACOL., vol. 316, 1996, pages 111 - 121
JAHNS, J. AM. COLL. CARDIOL., vol. 34, 1999, pages 1545 - 1551
JAHNS, J. AM. COLL. CARDIOL., vol. 36, 2000, pages 1280 - 1287
JAHNS, J. CLIN. INVEST., vol. 113, 2004, pages 1419 - 1429
JAHNS, TRENDS CARDIOVASC MED, vol. 16, 2006, pages 20 - 24
KATES S.; ALBERICIO F.: "Solid phase synthesis", 2000, CRC-PRESS
KHOYNEZHAD, EUR. J. HEART FAIL., vol. 9, 2007, pages 120 - 123
KÜHL, CIRCULATION, vol. 112, 2005, pages 1965 - 1970
LIMAS, AM. HEART J., vol. 123, 1992, pages 967 - 970
LIMAS, AM. J. CARDIOL, vol. 93, 2004, pages 1189 - 1191
LIMAS, CIRCULATION, vol. 95, 1997, pages 1979 - 1980
LIMAS, INT. J. CARDIOL., vol. 54, 1996, pages 113 - 116
LOHSE, CIRC. RES., vol. 93, 2003, pages 896 - 906
LUPPI, CIRCULATION, vol. 98, 1998, pages 777 - 785
LWATA, CIRC. RES., vol. 88, 2001, pages 578 - 586
LWATA, J. AM. COLL. CARDIOL., vol. 37, 2001, pages 418 - 424
MACLELLAN, NAT. MED., vol. 9, 2003, pages 1455 - 1456
MAEKAWA, CIRCULATION, vol. 115, 2007, pages 5 - 8
MAGNUSSON, CIRCULATION, vol. 89, 1994, pages 2760 - 2767
MAGNUSSON, INT. J. CARDIOL., vol. 54, 1996, pages 137 - 141
MAHRHOLDT, CIRCULATION, vol. 114, 2006, pages 1581 - 1590
MATSUI, AUTOIMMUNITY, vol. 21, 1995, pages 85 - 88
MATSUI, AUTOIMMUNITY, vol. 43, 2001, pages 217 - 220
MOBINI, AUTOIMMUNITY REV., vol. 3, 2004, pages 277 - 284
MORITA, J. CLIN. INVEST., vol. 115, 2005, pages 518 - 526
NEUMANN, J. AM. COLL. CARDIOL., vol. 16, 1990, pages 839 - 846
NIKOLAEV, J. AM. COLL. CARDIOL., vol. 50, 2007, pages 423 - 431
OKAZAKI, NAT. MED., vol. 9, 2003, pages 1477 - 1483
OKAZAKI, TRENDS MOI MED, vol. 11, 2005, pages 322 - 326
POHLNER, AM. J. CARDIOL., vol. 80, 1997, pages 1040 - 1045
RICHARDSON, CIRCULATION, vol. 93, 1996, pages 841 - 842
ROSE, IMMUNOL. TODAY, vol. 14, 1993, pages 426 - 430
ROSE, J. CLIN. INVEST., vol. 107, 2001, pages 943 - 944
SAMBROOK: "Molecular Cloning A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY
SCHULTHEISS, J. EXP. MED., vol. 168, 1988, pages 2102 - 2109
SCHULTHEISS, J. MOL. CELL CARDIOL., vol. 17, 1985, pages 603 - 617
SCHULZE, CARDIOVASC. RES., vol. 44, 1999, pages 91 - 100
SEWALD: "Peptides: Chemistry and Biology", 2002, WILLEY-VCH
SMULSKI, FASEB J., vol. 20, 2006, pages 1396 - 1406
STORK, AM. HEART J., vol. 152, 2006, pages 697 - 704
STRYER: "Biochemie", 1991, SPECTRUM AKAD. VERLAG
TIJSSEN: "Practice and theory of enzyme immunoassays", vol. 15, 1985
WALLUKAT, J. MOL. CELL CARDIOL., vol. 27, 1995, pages 397 - 406
WALLUKAT, N. ENGL. J. MED., vol. 347, 2002, pages 1806
WANG, BLOOD PRESSURE, vol. 3, 1996, pages 25 - 27
WARME, NATURE, 2008
WILLIAMS: "Chemical Approaches to the Synthesis of Peptides", 1997, CRC-PRESS
WITEBSKY, J. AM. MED. ASSOC., vol. 164, 1957, pages 1439 - 1447
WOODIWISS, CIRCULATION, vol. 103, 2001, pages 155 - 160
ZACCOLO M; POZZAN T., SCIENCE, vol. 295, 2002, pages 1711 - 1715

Also Published As

Publication number Publication date
HK1176363A1 (en) 2013-07-26
JP2010536356A (ja) 2010-12-02
CN101835793A (zh) 2010-09-15
CN101835793B (zh) 2014-04-23
PT2197900E (pt) 2012-10-24
US9062095B2 (en) 2015-06-23
CN103992406A (zh) 2014-08-20
EP2197900B9 (de) 2013-01-09
JP2013176369A (ja) 2013-09-09
EP2497777A3 (de) 2012-10-10
JP2013138679A (ja) 2013-07-18
EP2559698A3 (de) 2013-03-06
EP2559698B1 (de) 2014-11-12
DK2197900T3 (da) 2012-10-22
JP5248610B2 (ja) 2013-07-31
US20100209445A1 (en) 2010-08-19
HRP20120827T1 (hr) 2012-11-30
SI2197900T1 (sl) 2012-11-30
PL2197900T3 (pl) 2012-12-31
ES2391358T3 (es) 2012-11-23
EP2197900B1 (de) 2012-08-01
EP2197900A2 (de) 2010-06-23
EP2497777A2 (de) 2012-09-12
WO2009027063A2 (en) 2009-03-05
AU2008291296B2 (en) 2014-02-06
WO2009027063A3 (en) 2009-07-23
CA2697108A1 (en) 2009-03-05
CA2697108C (en) 2018-05-22
AU2008291296A1 (en) 2009-03-05

Similar Documents

Publication Publication Date Title
EP2197900B9 (de) Beta 1-adrenozeptor-antikörper inhibierende, mutierte, doppelt cyclisierte rezeptorpeptide
JP5788430B2 (ja) 抗β1アドレナリン受容体抗体を阻害する手段
EP2970417B1 (de) Bh4-stabilisierte peptide und verwendungen davon
DK1781315T3 (en) PEPTIDE INHIBITORS FOR MEDIATION OF stress responses
EP2391637A1 (de) Neue peptidhomologe zum inhibieren von beta1-adrenozeptor-antikörpern
US20150038673A1 (en) Mutant double cyclized receptor peptides inhibiting beta1-adrenoceptor antibodies
AU2013205477A1 (en) Mutant double cyclized receptor peptides inhibiting beta1-adrenoceptor antibodies
CA2234723A1 (en) Generating d-peptides: methods and compositions
AU2013200517A1 (en) Means for the inhibition of anti-beta1-adrenergic receptor antibodies

Legal Events

Date Code Title Description
PUAL Search report despatched

Free format text: ORIGINAL CODE: 0009013

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AC Divisional application: reference to earlier application

Ref document number: 2197900

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

AK Designated contracting states

Kind code of ref document: A3

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

RIC1 Information provided on ipc code assigned before grant

Ipc: G01N 33/68 20060101ALI20130131BHEP

Ipc: A61K 38/12 20060101ALI20130131BHEP

Ipc: C07K 14/705 20060101ALI20130131BHEP

Ipc: C07K 7/64 20060101AFI20130131BHEP

Ipc: A61K 38/16 20060101ALI20130131BHEP

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1176363

Country of ref document: HK

17P Request for examination filed

Effective date: 20130905

RBV Designated contracting states (corrected)

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

17Q First examination report despatched

Effective date: 20131003

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20140605

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AC Divisional application: reference to earlier application

Ref document number: 2197900

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 695697

Country of ref document: AT

Kind code of ref document: T

Effective date: 20141115

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602008035406

Country of ref document: DE

Effective date: 20141224

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: VOSSIUS AND PARTNER, CH

REG Reference to a national code

Ref country code: NL

Ref legal event code: VDEP

Effective date: 20141112

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 695697

Country of ref document: AT

Kind code of ref document: T

Effective date: 20141112

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150312

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150312

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150212

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150213

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

REG Reference to a national code

Ref country code: HK

Ref legal event code: GR

Ref document number: 1176363

Country of ref document: HK

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602008035406

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20150813

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150822

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

REG Reference to a national code

Ref country code: CH

Ref legal event code: PFA

Owner name: JULIUS-MAXIMILIANS-UNIVERSITAET WUERZBURG, DE

Free format text: FORMER OWNER: JULIUS-MAXIMILIANS-UNIVERSITAET WUERZBURG, DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150822

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20160826

Year of fee payment: 9

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20080822

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 10

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141112

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20170822

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170822

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20180817

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20180827

Year of fee payment: 11

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190831

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190831

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190831

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20230830

Year of fee payment: 16